Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Mol Cell ; 83(19): 3546-3557.e8, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802027

ABSTRACT

Nonstructural protein 1 (Nsp1) produced by coronaviruses inhibits host protein synthesis. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Nsp1 C-terminal domain was shown to bind the ribosomal mRNA channel to inhibit translation, but it is unclear whether this mechanism is broadly used by coronaviruses, whether the Nsp1 N-terminal domain binds the ribosome, or how Nsp1 allows viral RNAs to be translated. Here, we investigated Nsp1 from SARS-CoV-2, Middle East respiratory syndrome coronavirus (MERS-CoV), and Bat-Hp-CoV coronaviruses using structural, biophysical, and biochemical experiments, revealing a conserved role for the C-terminal domain. Additionally, the N-terminal domain of Bat-Hp-CoV Nsp1 binds to the decoding center of the 40S subunit, where it would prevent mRNA and eIF1A accommodation. Structure-based experiments demonstrated the importance of decoding center interactions in all three coronaviruses and showed that the same regions of Nsp1 are necessary for the selective translation of viral RNAs. Our results provide a mechanistic framework to understand how Nsp1 controls preferential translation of viral RNAs.


Subject(s)
COVID-19 , Chiroptera , Animals , Chiroptera/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Domains , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
2.
Mol Cell ; 73(3): 490-504.e6, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30581145

ABSTRACT

Fused in sarcoma (FUS) is an RNA binding protein involved in regulating many aspects of RNA processing and linked to several neurodegenerative diseases. Transcriptomics studies indicate that FUS binds a large variety of RNA motifs, suggesting that FUS RNA binding might be quite complex. Here, we present solution structures of FUS zinc finger (ZnF) and RNA recognition motif (RRM) domains bound to RNA. These structures show a bipartite binding mode of FUS comprising of sequence-specific recognition of a NGGU motif via the ZnF and an unusual shape recognition of a stem-loop RNA via the RRM. In addition, sequence-independent interactions via the RGG repeats significantly increase binding affinity and promote destabilization of structured RNA conformation, enabling additional binding. We further show that disruption of the RRM and ZnF domains abolishes FUS function in splicing. Altogether, our results rationalize why deciphering the RNA binding mode of FUS has been so challenging.


Subject(s)
RNA-Binding Protein FUS/chemistry , RNA/chemistry , Binding Sites , HeLa Cells , Humans , Models, Molecular , Nucleotide Motifs , Protein Binding , Protein Interaction Domains and Motifs , RNA/genetics , RNA/metabolism , RNA Recognition Motif , RNA Splicing , RNA Stability , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Structure-Activity Relationship , Zinc Fingers
3.
Trends Biochem Sci ; 47(11): 921-935, 2022 11.
Article in English | MEDLINE | ID: mdl-35780009

ABSTRACT

The term 'nonsense-mediated mRNA decay' (NMD) was initially coined to describe the translation-dependent degradation of mRNAs harboring premature termination codons (PTCs), but it is meanwhile known that NMD also targets many canonical mRNAs with numerous biological implications. The molecular mechanisms determining on which RNAs NMD ensues are only partially understood. Considering the broad range of NMD-sensitive RNAs and the variable degrees of their degradation, we highlight here the hallmarks of mammalian NMD and point out open questions. We review the links between NMD and disease by summarizing the role of NMD in cancer, neurodegeneration, and viral infections. Finally, we describe strategies to modulate NMD activity and specificity as potential therapeutic approaches for various diseases.


Subject(s)
Codon, Nonsense , Nonsense Mediated mRNA Decay , Animals , Mammals , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Nucleic Acids Res ; 50(11): 6300-6312, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35687109

ABSTRACT

Heterogenous nuclear ribonucleoproteins (hnRNPs) are abundant proteins implicated in various steps of RNA processing that assemble on nuclear RNA into larger complexes termed 40S hnRNP particles. Despite their initial discovery 55 years ago, our understanding of these intriguing macromolecular assemblies remains limited. Here, we report the biochemical purification of native 40S hnRNP particles and the determination of their complete protein composition by label-free quantitative mass spectrometry, identifying A-group and C-group hnRNPs as the major protein constituents. Isolated 40S hnRNP particles dissociate upon RNA digestion and can be reconstituted in vitro on defined RNAs in the presence of the individual protein components, demonstrating a scaffolding role for RNA in nucleating particle formation. Finally, we revealed their nanometer scale, condensate-like nature, promoted by intrinsically disordered regions of A-group hnRNPs. Collectively, we identify nuclear 40S hnRNP particles as novel dynamic biomolecular condensates.


Subject(s)
Biomolecular Condensates , Heterogeneous-Nuclear Ribonucleoproteins , Cell Nucleus/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA/metabolism
5.
PLoS Pathog ; 17(5): e1009603, 2021 05.
Article in English | MEDLINE | ID: mdl-34019569

ABSTRACT

The positive-sense, single-stranded RNA alphaviruses pose a potential epidemic threat. Understanding the complex interactions between the viral and the host cell proteins is crucial for elucidating the mechanisms underlying successful virus replication strategies and for developing specific antiviral interventions. Here we present the first comprehensive protein-protein interaction map between the proteins of Semliki Forest Virus (SFV), a mosquito-borne member of the alphaviruses, and host cell proteins. Among the many identified cellular interactors of SFV proteins, the enrichment of factors involved in translation and nonsense-mediated mRNA decay (NMD) was striking, reflecting the virus' hijacking of the translation machinery and indicating viral countermeasures for escaping NMD by inhibiting NMD at later time points during the infectious cycle. In addition to observing a general inhibition of NMD about 4 hours post infection, we also demonstrate that transient expression of the SFV capsid protein is sufficient to inhibit NMD in cells, suggesting that the massive production of capsid protein during the SFV reproduction cycle is responsible for NMD inhibition.


Subject(s)
Alphavirus Infections/virology , Capsid Proteins/metabolism , Host-Pathogen Interactions , Nonsense Mediated mRNA Decay/genetics , Semliki forest virus/physiology , Capsid Proteins/genetics , HeLa Cells , Humans , Semliki forest virus/genetics , Virus Replication
6.
Nucleic Acids Res ; 49(14): 8261-8276, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34232997

ABSTRACT

Newly synthesized mRNA is translated during its export through the nuclear pore complex, when its 5'-cap structure is still bound by the nuclear cap-binding complex (CBC), a heterodimer of cap-binding protein (CBP) 80 and CBP20. Despite its critical role in mRNA surveillance, the mechanism by which CBC-dependent translation (CT) is regulated remains unknown. Here, we demonstrate that the CT initiation factor (CTIF) is tethered in a translationally incompetent manner to the perinuclear region by the DEAD-box helicase 19B (DDX19B). DDX19B hands over CTIF to CBP80, which is associated with the 5'-cap of a newly exported mRNA. The resulting CBP80-CTIF complex then initiates CT in the perinuclear region. We also show that impeding the interaction between CTIF and DDX19B leads to uncontrolled CT throughout the cytosol, consequently dysregulating nonsense-mediated mRNA decay. Altogether, our data provide molecular evidence supporting the importance of tight control of local translation in the perinuclear region.


Subject(s)
DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factors/genetics , Nuclear Cap-Binding Protein Complex/genetics , Nucleocytoplasmic Transport Proteins/genetics , Protein Biosynthesis , Cytoplasm/genetics , HeLa Cells , Humans , Nonsense Mediated mRNA Decay/genetics , Protein Interaction Maps/genetics , RNA Cap-Binding Proteins/genetics , RNA, Messenger/genetics
7.
Nucleic Acids Res ; 49(13): 7713-7731, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34233002

ABSTRACT

Liquid-liquid phase separation (LLPS) of proteins and RNAs has emerged as the driving force underlying the formation of membrane-less organelles. Such biomolecular condensates have various biological functions and have been linked to disease. The protein Fused in Sarcoma (FUS) undergoes LLPS and mutations in FUS have been causally linked to the motor neuron disease Amyotrophic Lateral Sclerosis (ALS-FUS). LLPS followed by aggregation of cytoplasmic FUS has been proposed to be a crucial disease mechanism. However, it is currently unclear how LLPS impacts the behaviour of FUS in cells, e.g. its interactome. Hence, we developed a method allowing for the purification of LLPS FUS-containing droplets from cell lysates. We observe substantial alterations in the interactome, depending on its biophysical state. While non-LLPS FUS interacts mainly with factors involved in pre-mRNA processing, LLPS FUS predominantly binds to proteins involved in chromatin remodelling and DNA damage repair. Interestingly, also mitochondrial factors are strongly enriched with LLPS FUS, providing a potential explanation for the observed changes in mitochondrial gene expression in mouse models of ALS-FUS. In summary, we present a methodology to investigate the interactomes of phase separating proteins and provide evidence that LLPS shapes the FUS interactome with implications for function and disease.


Subject(s)
RNA-Binding Protein FUS/metabolism , Cell Nucleus/metabolism , Chromatin/metabolism , Cytoplasm/metabolism , Cytoplasmic Granules/metabolism , HEK293 Cells , HeLa Cells , Humans , Protein Interaction Mapping , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/isolation & purification
8.
RNA Biol ; 19(1): 609-621, 2022.
Article in English | MEDLINE | ID: mdl-35491909

ABSTRACT

Cells of metazoans respond to internal and external stressors by activating stress response pathways that aim for re-establishing cellular homoeostasis or, if this cannot be achieved, triggering programmed cell death. Problems during translation, arising from defective mRNAs, tRNAs, ribosomes or protein misfolding, can activate stress response pathways as well as mRNA surveillance and ribosome quality control programs. Recently, ribosome collisions have emerged as a central signal for translational stress and shown to elicit different stress responses. Here, we review our current knowledge about the intricate mutual connections between ribosome collisions, stress response pathways and mRNA surveillance. A central factor connecting the sensing of collided ribosomes with degradation of the nascent polypeptides, dissociation of the stalled ribosomes and degradation of the mRNA by no-go or non-stop decay is the E3-ligase ZNF598. We tested whether ZNF598 also plays a role in nonsense-mediated mRNA decay (NMD) but found that it is dispensable for this translation termination-associated mRNA surveillance pathway, which in combination with other recent data argues against stable ribosome stalling at termination codons being the NMD-triggering signal.


Subject(s)
Insurance , Ribosomes , Nonsense Mediated mRNA Decay , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
9.
RNA Biol ; 19(1): 78-88, 2022 01.
Article in English | MEDLINE | ID: mdl-34965175

ABSTRACT

Protein synthesis is a central process in gene expression and the development of efficient in vitro translation systems has been the focus of scientific efforts for decades. The production of translation-competent lysates originating from human cells or tissues remains challenging, mainly due to the variability of cell lysis conditions. Here we present a robust and fast method based on dual centrifugation that allows for detergent-free cell lysis under controlled mechanical forces. We optimized the lysate preparation to yield cytoplasm-enriched extracts from human cells that efficiently translate mRNAs in a cap-dependent as well as in an IRES-mediated way. Reduction of the phosphorylation state of eIF2α using recombinant GADD34 and 2-aminopurine considerably boosts the protein output, reinforcing the potential of this method to produce recombinant proteins from human lysates.


Subject(s)
Cell Fractionation , Cell-Free System , Centrifugation , In Vitro Techniques , Protein Biosynthesis , Cell Fractionation/methods , Centrifugation/methods , Genes, Reporter , HeLa Cells , Humans , RNA, Messenger/genetics , Subcellular Fractions
10.
Nucleic Acids Res ; 48(18): 10259-10279, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32941650

ABSTRACT

To gain insight into the mechanistic link between translation termination and nonsense-mediated mRNA decay (NMD), we depleted the ribosome recycling factor ABCE1 in human cells, resulting in an upregulation of NMD-sensitive mRNAs. Suppression of NMD on these mRNAs occurs prior to their SMG6-mediated endonucleolytic cleavage. ABCE1 depletion caused ribosome stalling at termination codons (TCs) and increased ribosome occupancy in 3' UTRs, implying enhanced TC readthrough. ABCE1 knockdown indeed increased the rate of readthrough and continuation of translation in different reading frames, providing a possible explanation for the observed NMD inhibition, since enhanced readthrough displaces NMD activating proteins from the 3' UTR. Our results indicate that stalling at TCs triggers ribosome collisions and activates ribosome quality control. Collectively, we show that improper translation termination can lead to readthrough of the TC, presumably due to ribosome collisions pushing the stalled ribosomes into the 3' UTR, where it can resume translation in-frame as well as out-of-frame.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Codon, Terminator/genetics , Nonsense Mediated mRNA Decay/genetics , Telomerase/genetics , 3' Untranslated Regions/genetics , Frameshifting, Ribosomal/genetics , Humans , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Ribosomes/genetics
11.
Nucleic Acids Res ; 48(12): 6889-6905, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32479602

ABSTRACT

Mutations in the RNA-binding protein FUS cause amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease. FUS plays a role in numerous aspects of RNA metabolism, including mRNA splicing. However, the impact of ALS-causative mutations on splicing has not been fully characterized, as most disease models have been based on overexpressing mutant FUS, which will alter RNA processing due to FUS autoregulation. We and others have recently created knockin models that overcome the overexpression problem, and have generated high depth RNA-sequencing on FUS mutants in parallel to FUS knockout, allowing us to compare mutation-induced changes to genuine loss of function. We find that FUS-ALS mutations induce a widespread loss of function on expression and splicing. Specifically, we find that mutant FUS directly alters intron retention levels in RNA-binding proteins. Moreover, we identify an intron retention event in FUS itself that is associated with its autoregulation. Altered FUS levels have been linked to disease, and we show here that this novel autoregulation mechanism is altered by FUS mutations. Crucially, we also observe this phenomenon in other genetic forms of ALS, including those caused by TDP-43, VCP and SOD1 mutations, supporting the concept that multiple ALS genes interact in a regulatory network.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Homeostasis/genetics , RNA-Binding Protein FUS/genetics , Animals , Cytoplasm/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Introns/genetics , Loss of Function Mutation , Mice , Mice, Knockout , Mutation/genetics , RNA Splicing/genetics , Superoxide Dismutase-1/genetics , Valosin Containing Protein/genetics
12.
Semin Cell Dev Biol ; 75: 78-87, 2018 03.
Article in English | MEDLINE | ID: mdl-28866327

ABSTRACT

Nonsense-mediated mRNA decay (NMD) has traditionally been described as a quality control system that rids cells of aberrant mRNAs with crippled protein coding potential. However, transcriptome-wide profiling of NMD deficient cells identified a plethora of seemingly intact mRNAs coding for functional proteins as NMD targets. This led to the view that NMD constitutes an additional post-transcriptional layer of gene expression control involved in the regulation of many different biological pathways. Here, we review our current knowledge about the role of NMD in embryonic development and tissue-specific cell differentiation. We further summarize how NMD contributes to balancing of the integrated stress response and to cellular homeostasis of splicing regulators and NMD factors through auto-regulatory feedback loops. In addition, we discuss recent evidence that suggests a role for NMD as an innate immune response against several viruses. Altogether, NMD appears to play an important role in a broad spectrum of biological pathways, many of which still remain to be discovered.


Subject(s)
Gene Expression Regulation , Homeostasis/genetics , Nonsense Mediated mRNA Decay , RNA, Messenger/genetics , Animals , Cell Differentiation/genetics , Embryonic Development/genetics , Humans , Immunity, Innate/genetics , Quality Control , RNA, Messenger/metabolism
13.
EMBO J ; 35(14): 1504-21, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27252488

ABSTRACT

Fused in sarcoma (FUS) is a ubiquitously expressed RNA-binding protein proposed to function in various RNA metabolic pathways, including transcription regulation, pre-mRNA splicing, RNA transport and microRNA processing. Mutations in the FUS gene were identified in patients with amyotrophic lateral sclerosis (ALS), but the pathomechanisms by which these mutations cause ALS are not known. Here, we show that FUS interacts with the minor spliceosome constituent U11 snRNP, binds preferentially to minor introns and directly regulates their removal. Furthermore, a FUS knockout in neuroblastoma cells strongly disturbs the splicing of minor intron-containing mRNAs, among them mRNAs required for action potential transmission and for functional spinal motor units. Moreover, an ALS-associated FUS mutant that forms cytoplasmic aggregates inhibits splicing of minor introns by trapping U11 and U12 snRNAs in these aggregates. Collectively, our findings suggest a possible pathomechanism for ALS in which mutated FUS inhibits correct splicing of minor introns in mRNAs encoding proteins required for motor neuron survival.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Introns , Mutant Proteins/genetics , Mutant Proteins/metabolism , RNA Splicing , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Cell Line , Humans
14.
RNA ; 24(4): 557-573, 2018 04.
Article in English | MEDLINE | ID: mdl-29348139

ABSTRACT

The term "nonsense-mediated mRNA decay" (NMD) originally described the degradation of mRNAs with premature translation-termination codons (PTCs), but its meaning has recently been extended to be a translation-dependent post-transcriptional regulator of gene expression affecting 3%-10% of all mRNAs. The degradation of NMD target mRNAs involves both exonucleolytic and endonucleolytic pathways in mammalian cells. While the latter is mediated by the endonuclease SMG6, the former pathway has been reported to require a complex of SMG5-SMG7 or SMG5-PNRC2 binding to UPF1. However, the existence, dominance, and mechanistic details of these exonucleolytic pathways are divisive. Therefore, we have investigated the possible exonucleolytic modes of mRNA decay in NMD by examining the roles of UPF1, SMG5, SMG7, and PNRC2 using a combination of functional assays and interaction mapping. Confirming previous work, we detected an interaction between SMG5 and SMG7 and also a functional need for this complex in NMD. In contrast, we found no evidence for the existence of a physical or functional interaction between SMG5 and PNRC2. Instead, we show that UPF1 interacts with PNRC2 and that it triggers 5'-3' exonucleolytic decay of reporter transcripts in tethering assays. PNRC2 interacts mainly with decapping factors and its knockdown does not affect the RNA levels of NMD reporters. We conclude that PNRC2 is probably an important mRNA decapping factor but that it does not appear to be required for NMD.


Subject(s)
Carrier Proteins/metabolism , Nonsense Mediated mRNA Decay/genetics , RNA Helicases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Codon, Nonsense/genetics , Gene Expression Regulation/genetics , HeLa Cells , Humans , Protein Binding/genetics , RNA Helicases/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Trans-Activators/genetics , Two-Hybrid System Techniques
15.
RNA ; 23(2): 189-201, 2017 02.
Article in English | MEDLINE | ID: mdl-27864472

ABSTRACT

Besides degrading aberrant mRNAs that harbor a premature translation termination codon (PTC), nonsense-mediated mRNA decay (NMD) also targets many seemingly "normal" mRNAs that encode for full-length proteins. To identify a bona fide set of such endogenous NMD targets in human cells, we applied a meta-analysis approach in which we combined transcriptome profiling of knockdowns and rescues of the three NMD factors UPF1, SMG6, and SMG7. We provide evidence that this combinatorial approach identifies NMD-targeted transcripts more reliably than previous attempts that focused on inactivation of single NMD factors. Our data revealed that SMG6 and SMG7 act on essentially the same transcripts, indicating extensive redundancy between the endo- and exonucleolytic decay routes. Besides mRNAs, we also identified as NMD targets many long noncoding RNAs as well as miRNA and snoRNA host genes. The NMD target feature with the most predictive value is an intron in the 3' UTR, followed by the presence of upstream open reading frames (uORFs) and long 3' UTRs. Furthermore, the 3' UTRs of NMD-targeted transcripts tend to have an increased GC content and to be phylogenetically less conserved when compared to 3' UTRs of NMD insensitive transcripts.


Subject(s)
Carrier Proteins/metabolism , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism , Telomerase/metabolism , Trans-Activators/metabolism , Transcriptome , Base Composition , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Codon, Nonsense , Gene Expression , HeLa Cells , Humans , Introns , MicroRNAs/chemistry , MicroRNAs/metabolism , Protein Binding , RNA Helicases , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , RNA, Messenger/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/metabolism , Telomerase/antagonists & inhibitors , Telomerase/genetics , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
16.
Chimia (Aarau) ; 73(6): 374-379, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31118119

ABSTRACT

Eukaryotic cells have evolved a number of survival tactics and quality control pathways that are able to counter intrinsic error-prone mechanisms and allow for maintenance of cellular homeostasis in the face of external stresses. This review will discuss the role of two cellular eukaryotic processes that are vital for maintenance of cellular homeostasis - 1) the nonsense-mediated mRNA decay (NMD) pathway and 2) the transient formation of stress granules (SG) - and explore the current literature on their roles in antiviral defence. Within the NCCR RNA & Disease, the laboratories of Proffs. O. Mühlemann and Volker Thiel teamed up to unravel the roles of NMD and SGs, and their interconnections in defending cells from alphavirus and coronavirus infections.


Subject(s)
Nonsense Mediated mRNA Decay , Antiviral Agents , RNA
18.
Nucleic Acids Res ; 42(14): 9217-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25053839

ABSTRACT

Eukaryotic mRNAs with premature translation-termination codons (PTCs) are recognized and eliminated by nonsense-mediated mRNA decay (NMD). NMD substrates can be degraded by different routes that all require phosphorylated UPF1 (P-UPF1) as a starting point. The endonuclease SMG6, which cleaves mRNA near the PTC, is one of the three known NMD factors thought to be recruited to nonsense mRNAs via an interaction with P-UPF1, leading to eventual mRNA degradation. By artificial tethering of SMG6 and mutants thereof to a reporter mRNA combined with knockdowns of various NMD factors, we demonstrate that besides its endonucleolytic activity, SMG6 also requires UPF1 and SMG1 to reduce reporter mRNA levels. Using in vivo and in vitro approaches, we further document that SMG6 and the unique stalk region of the UPF1 helicase domain, along with a contribution from the SQ domain, form a novel interaction and we also show that this region of the UPF1 helicase domain is critical for SMG6 function and NMD. Our results show that this interaction is required for NMD and for the capability of tethered SMG6 to degrade its bound RNA, suggesting that it contributes to the intricate regulation of UPF1 and SMG6 enzymatic activities.


Subject(s)
Nonsense Mediated mRNA Decay , Telomerase/metabolism , Trans-Activators/metabolism , Binding Sites , HeLa Cells , Humans , Mutation , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , RNA Helicases , Telomerase/chemistry , Trans-Activators/chemistry , Trans-Activators/genetics
19.
Trends Genet ; 28(2): 70-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154474

ABSTRACT

The accuracy of eukaryotic gene expression relies on efficient quality control (QC). Most steps in the gene expression pathway en route from transcription to translation are error-prone and QC systems have evolved to utilise many of these biochemical processes as checkpoints to monitor the production or function of mRNA-protein particles (mRNPs). Mechanistically, such evaluation of mRNP fitness is based on competition between the opposing activities of mRNP biogenesis and/or function and mRNP turnover. In fact, quite subtle alteration of any parameter can tip the balance between mRNP persistence and degradation and, therefore, QC checkpoints also comprise perfect opportunities for controlling cellular levels of individual or even entire families of transcripts. From this perspective, QC and gene regulation represent two outcomes of the same molecular process.


Subject(s)
Gene Expression Regulation , Ribonucleoproteins/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Protein Biosynthesis , RNA Stability
20.
RNA ; 19(10): 1432-48, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23962664

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is a eukaryotic post-transcriptional gene regulation mechanism that eliminates mRNAs with the termination codon (TC) located in an unfavorable environment for efficient translation termination. The best-studied NMD-targeted mRNAs contain premature termination codons (PTCs); however, NMD regulates even many physiological mRNAs. An exon-junction complex (EJC) located downstream from a TC acts as an NMD-enhancing signal, but is not generally required for NMD. Here, we compared these "EJC-enhanced" and "EJC-independent" modes of NMD with regard to their requirement for seven known NMD factors in human cells using two well-characterized NMD reporter genes (immunoglobulin µ and ß-Globin) with or without an intron downstream from the PTC. We show that both NMD modes depend on UPF1 and SMG1, but detected transcript-specific differences with respect to the requirement for UPF2 and UPF3b, consistent with previously reported UPF2- and UPF3-independent branches of NMD. In addition and contrary to expectation, a higher sensitivity of EJC-independent NMD to reduced UPF2 and UPF3b concentrations was observed. Our data further revealed a redundancy of the endo- and exonucleolytic mRNA degradation pathways in both modes of NMD. Moreover, the relative contributions of both decay pathways differed between the reporters, with PTC-containing immunoglobulin µ transcripts being preferentially subjected to SMG6-mediated endonucleolytic cleavage, whereas ß-Globin transcripts were predominantly degraded by the SMG5/SMG7-dependent pathway. Overall, the surprising heterogeneity observed with only two NMD reporter pairs suggests the existence of several mechanistically distinct branches of NMD in human cells.


Subject(s)
Codon, Nonsense/genetics , Exons/genetics , Gene Expression Regulation , Nonsense Mediated mRNA Decay/genetics , RNA Stability/genetics , RNA, Messenger/genetics , Blotting, Western , HeLa Cells , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , RNA Helicases , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL