Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
EMBO J ; 41(10): e109191, 2022 05 16.
Article in English | MEDLINE | ID: mdl-35451084

ABSTRACT

The paralogous human proteins UPF3A and UPF3B are involved in recognizing mRNAs targeted by nonsense-mediated mRNA decay (NMD). UPF3B has been demonstrated to support NMD, presumably by bridging an exon junction complex (EJC) to the NMD factor UPF2. The role of UPF3A has been described either as a weak NMD activator or an NMD inhibitor. Here, we present a comprehensive functional analysis of UPF3A and UPF3B in human cells using combinatory experimental approaches. Overexpression or knockout of UPF3A as well as knockout of UPF3B did not substantially change global NMD activity. In contrast, the co-depletion of UPF3A and UPF3B resulted in a marked NMD inhibition and a transcriptome-wide upregulation of NMD substrates, demonstrating a functional redundancy between both NMD factors. In rescue experiments, UPF2 or EJC binding-deficient UPF3B largely retained NMD activity. However, combinations of different mutants, including deletion of the middle domain, showed additive or synergistic effects and therefore failed to maintain NMD. Collectively, UPF3A and UPF3B emerge as fault-tolerant, functionally redundant NMD activators in human cells.


Subject(s)
Nonsense Mediated mRNA Decay , RNA-Binding Proteins , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Transcriptome
2.
Mol Cell ; 72(3): 482-495.e7, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388410

ABSTRACT

Productive splicing of human precursor messenger RNAs (pre-mRNAs) requires the correct selection of authentic splice sites (SS) from the large pool of potential SS. Although SS consensus sequence and splicing regulatory proteins are known to influence SS usage, the mechanisms ensuring the effective suppression of cryptic SS are insufficiently explored. Here, we find that many aberrant exonic SS are efficiently silenced by the exon junction complex (EJC), a multi-protein complex that is deposited on spliced mRNA near the exon-exon junction. Upon depletion of EJC proteins, cryptic SS are de-repressed, leading to the mis-splicing of a broad set of mRNAs. Mechanistically, the EJC-mediated recruitment of the splicing regulator RNPS1 inhibits cryptic 5'SS usage, while the deposition of the EJC core directly masks reconstituted 3'SS, thereby precluding transcript disintegration. Thus, the EJC protects the transcriptome of mammalian cells from inadvertent loss of exonic sequences and safeguards the expression of intact, full-length mRNAs.


Subject(s)
Alternative Splicing/physiology , Exons/physiology , RNA Splice Sites/physiology , Consensus Sequence/genetics , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , HeLa Cells , Humans , Introns , RNA Precursors/physiology , RNA Splicing/physiology , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Transcriptome/genetics
3.
Trends Genet ; 37(4): 355-372, 2021 04.
Article in English | MEDLINE | ID: mdl-33203572

ABSTRACT

Splicing of precursor mRNAs (pre-mRNA) is an important step during eukaryotic gene expression. The identification of the actual splice sites and the proper removal of introns are essential for the production of the desired mRNA isoforms and their encoded proteins. While the basic mechanisms of splicing regulation are well understood, recent work has uncovered a growing number of noncanonical splicing mechanisms that play key roles in the regulation of gene expression. In this review, we summarize the current principles of splicing regulation, including the impact of cis and trans regulatory elements, as well as the influence of chromatin structure, transcription, and RNA modifications. We further discuss the recent development of emerging splicing mechanisms, such as recursive and back splicing, and their impact on gene expression.


Subject(s)
Alternative Splicing/genetics , Eukaryota/genetics , RNA Precursors/genetics , RNA Splicing/genetics , Humans , Introns/genetics , RNA, Messenger/genetics
4.
Cell ; 137(3): 536-48, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19410547

ABSTRACT

Exon junction complexes (EJCs) are deposited onto mRNAs during splicing, serve as positional landmarks for the intron exon structure of genes, and direct posttranscriptional processes in the cytoplasm. EJC removal and recycling by translation are ill understood and have been attributed to ribosomal passage. This work identifies the ribosome-associated protein PYM as an EJC disassembly factor and defines its mechanism of function. Whereas EJC assembly intermediates are resistant to PYM, fully assembled EJCs are dissociated from spliced mRNAs by PYM. This disassembly involves PYM binding to the EJC proteins MAGOH-Y14. PYM overexpression in cells disrupts EJC association with spliced mRNA and inhibits nonsense-mediated mRNA decay. In cells depleted of PYM, EJCs accumulate on spliced mRNAs and EJC protein recycling is impaired. Hence, PYM is an EJC disassembly factor that acts both in vitro and in living cells, and that antagonizes important EJC functions.


Subject(s)
Carrier Proteins , Exons , RNA Splicing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Humans , Introns , Protein Binding , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism
5.
Nucleic Acids Res ; 50(10): 5899-5918, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35640609

ABSTRACT

The exon junction complex (EJC) is an RNA-binding multi-protein complex with critical functions in post-transcriptional gene regulation. It is deposited on the mRNA during splicing and regulates diverse processes including pre-mRNA splicing and nonsense-mediated mRNA decay (NMD) via various interacting proteins. The peripheral EJC-binding protein RNPS1 was reported to serve two insufficiently characterized functions: suppressing mis-splicing of cryptic splice sites and activating NMD in the cytoplasm. The analysis of transcriptome-wide effects of EJC and RNPS1 knockdowns in different human cell lines supports the conclusion that RNPS1 can moderately influence NMD activity, but is not a globally essential NMD factor. However, numerous aberrant splicing events strongly suggest that the main function of RNPS1 is splicing regulation. Rescue analyses revealed that the RRM and C-terminal domain of RNPS1 both contribute partially to regulate RNPS1-dependent splicing events. We defined the RNPS1 core interactome using complementary immunoprecipitations and proximity labeling, which identified interactions with splicing-regulatory factors that are dependent on the C-terminus or the RRM domain of RNPS1. Thus, RNPS1 emerges as a multifunctional splicing regulator that promotes correct and efficient splicing of different vulnerable splicing events via the formation of diverse splicing-promoting complexes.


Subject(s)
Ribonucleoproteins , Transcriptome , Exons , Humans , RNA Splicing/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
6.
RNA ; 2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33906975

ABSTRACT

The current ecosystem of single cell RNA-seq platforms is rapidly expanding, but robust solutions for single cell and single molecule full- length RNA sequencing are virtually absent. A high-throughput solution that covers all aspects is necessary to study the complex life of mRNA on the single cell level. The Nanopore platform offers long read sequencing and can be integrated with the popular single cell sequencing method on the 10x Chromium platform. However, the high error-rate of Nanopore reads poses a challenge in downstream processing (e.g. for cell barcode assignment). We propose a solution to this particular problem by using a hybrid sequencing approach on Nanopore and Illumina platforms. Our software ScNapBar enables cell barcode assignment with high accuracy, especially if sequencing satura- tion is low. ScNapBar uses unique molecular identifier (UMI) or Naive Bayes probabilistic approaches in the barcode assignment, depending on the available Illumina sequencing depth. We have benchmarked the two approaches on simulated and real Nanopore datasets. We further applied ScNapBar to pools of cells with an active or a silenced non-sense mediated RNA decay pathway. Our Nanopore read assignment distinguishes the respective cell populations and reveals characteristic nonsense-mediated mRNA decay events depending on cell status.

7.
Nucleic Acids Res ; 49(3): 1688-1707, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33444449

ABSTRACT

Pre-mRNA splicing catalyzed by the spliceosome represents a critical step in the regulation of gene expression contributing to transcriptome and proteome diversity. The spliceosome consists of five small nuclear ribonucleoprotein particles (snRNPs), the biogenesis of which remains only partially understood. Here we define the evolutionarily conserved protein Ecdysoneless (Ecd) as a critical regulator of U5 snRNP assembly and Prp8 stability. Combining Drosophila genetics with proteomic approaches, we demonstrate the Ecd requirement for the maintenance of adult healthspan and lifespan and identify the Sm ring protein SmD3 as a novel interaction partner of Ecd. We show that the predominant task of Ecd is to deliver Prp8 to the emerging U5 snRNPs in the cytoplasm. Ecd deficiency, on the other hand, leads to reduced Prp8 protein levels and compromised U5 snRNP biogenesis, causing loss of splicing fidelity and transcriptome integrity. Based on our findings, we propose that Ecd chaperones Prp8 to the forming U5 snRNP allowing completion of the cytoplasmic part of the U5 snRNP biogenesis pathway necessary to meet the cellular demand for functional spliceosomes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , RNA Splicing Factors/metabolism , Ribonucleoprotein, U5 Small Nuclear/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Mutation , Protein Stability , RNA Splicing , Transcriptome
8.
Nucleic Acids Res ; 48(15): 8626-8644, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32621609

ABSTRACT

The exon junction complex (EJC) is an essential constituent and regulator of spliced messenger ribonucleoprotein particles (mRNPs) in metazoans. As a core component of the EJC, CASC3 was described to be pivotal for EJC-dependent nuclear and cytoplasmic processes. However, recent evidence suggests that CASC3 functions differently from other EJC core proteins. Here, we have established human CASC3 knockout cell lines to elucidate the cellular role of CASC3. In the knockout cells, overall EJC composition and EJC-dependent splicing are unchanged. A transcriptome-wide analysis reveals that hundreds of mRNA isoforms targeted by nonsense-mediated decay (NMD) are upregulated. Mechanistically, recruiting CASC3 to reporter mRNAs by direct tethering or via binding to the EJC stimulates mRNA decay and endonucleolytic cleavage at the termination codon. Building on existing EJC-NMD models, we propose that CASC3 equips the EJC with the persisting ability to communicate with the NMD machinery in the cytoplasm. Collectively, our results characterize CASC3 as a peripheral EJC protein that tailors the transcriptome by promoting the degradation of EJC-dependent NMD substrates.


Subject(s)
Neoplasm Proteins/genetics , Nonsense Mediated mRNA Decay/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Transcriptome/genetics , Amino Acid Sequence/genetics , Cell Nucleus/genetics , Exons/genetics , Gene Knockout Techniques , Humans , RNA, Messenger/genetics , Ribonucleoproteins/genetics
9.
Trends Biochem Sci ; 42(5): 369-382, 2017 05.
Article in English | MEDLINE | ID: mdl-28268044

ABSTRACT

Eukaryotic cells determine the final protein output of their genetic program not only by controlling transcription but also by regulating the localization, translation and turnover rates of their mRNAs. Ultimately, the fate of any given mRNA is determined by the ensemble of all associated RNA-binding proteins (RBPs), non-coding RNAs and metabolites collectively known as the messenger ribonucleoprotein particle (mRNP). Although many mRNA-associated factors have been identified over the past years, little is known about the composition of individual mRNPs and the cooperation of their constituents. In this review we discuss recent progress that has been made on how this 'mRNP code' is established on individual transcripts and how it is interpreted during gene expression in eukaryotic cells.


Subject(s)
Protein Processing, Post-Translational/genetics , RNA/genetics , RNA/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Animals , Eukaryotic Cells/metabolism , Gene Expression Regulation/genetics , Humans
10.
Trends Genet ; 32(11): 724-735, 2016 11.
Article in English | MEDLINE | ID: mdl-27667727

ABSTRACT

The exon junction complex (EJC) is an RNA-binding protein complex that is assembled and deposited onto mRNAs during splicing. The EJC comprises four core components that bind to not only canonical sites upstream of exon-exon junctions, but also to noncanonical sites at other positions in exons. EJC-associated proteins are recruited by the EJC at different steps of gene expression to execute the multiple functions of the EJC. Recently, new insights have been obtained into how EJCs stimulate pre-mRNA splicing, and mRNA export, translation, and degradation. Furthermore, mutations in EJC core components were shown to result in severe disorders in humans, demonstrating the critical physiological role of the EJC. Hence, the EJC has been identified as an important player in post-transcriptional gene regulation in metazoans.


Subject(s)
Exons/genetics , Multiprotein Complexes/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Mutation , RNA Precursors/genetics , RNA Stability/genetics , Ribonucleoproteins/genetics
11.
RNA ; 23(8): 1224-1236, 2017 08.
Article in English | MEDLINE | ID: mdl-28461625

ABSTRACT

Nonsense-mediated mRNA decay (NMD) controls gene expression by eliminating mRNAs with premature or aberrant translation termination. Degradation of NMD substrates is initiated by the central NMD factor UPF1, which recruits the endonuclease SMG6 and the deadenylation-promoting SMG5/7 complex. The extent to which SMG5/7 and SMG6 contribute to the degradation of individual substrates and their regulation by UPF1 remains elusive. Here we map transcriptome-wide sites of SMG6-mediated endocleavage via 3' fragment capture and degradome sequencing. This reveals that endogenous transcripts can have NMD-eliciting features at various positions, including upstream open reading frames (uORFs), premature termination codons (PTCs), and long 3' UTRs. We find that NMD substrates with PTCs undergo constitutive SMG6-dependent endocleavage, rather than SMG7-dependent exonucleolytic decay. In contrast, the turnover of NMD substrates containing uORFs and long 3' UTRs involves both SMG6- and SMG7-dependent endo- and exonucleolytic decay, respectively. This suggests that the extent to which SMG6 and SMG7 degrade NMD substrates is determined by the mRNA architecture.


Subject(s)
Carrier Proteins/metabolism , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism , Telomerase/metabolism , Carrier Proteins/genetics , Codon, Nonsense , HeLa Cells , Humans , Open Reading Frames , RNA Helicases , RNA, Messenger/genetics , Telomerase/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
12.
Biochem Soc Trans ; 46(1): 153-161, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29351963

ABSTRACT

During splicing, the exon junction complex (EJC) is deposited upstream of exon-exon boundaries. The EJC and its peripheral bound proteins play an essential role in mediating mRNA export, translation and turnover. However, the exact sequence of EJC assembly and the involved factors during splicing remain elusive. Recently published structures of the human C* spliceosome clarified the position of the EJC at this phase of splicing and have given insight into previously unidentified interactions between the EJC and spliceosomal proteins. Here, these new observations are presented and the significance for EJC assembly is discussed. Furthermore, the vast landscape of EJC interacting proteins and their manifold functions are described. Finally, the factors involved in EJC disassembly and recycling are recapitulated. This review aims to integrate structural, biochemical and physiological data to obtain a comprehensive picture of EJC components during the lifetime of the EJC.


Subject(s)
Exons , Ribonucleoproteins/metabolism , Animals , Mammals , RNA Splicing , Ribonucleoproteins/genetics , Spliceosomes/metabolism
13.
Nucleic Acids Res ; 44(5): 2348-61, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26773052

ABSTRACT

The export of messenger RNAs (mRNAs) is the final of several nuclear posttranscriptional steps of gene expression. The formation of export-competent mRNPs involves the recruitment of export factors that are assumed to facilitate transport of the mature mRNAs. Using in vitro splicing assays, we show that a core set of export factors, including ALYREF, UAP56 and DDX39, readily associate with the spliced RNAs in an EJC (exon junction complex)- and cap-dependent manner. In order to elucidate how ALYREF and other export adaptors mediate mRNA export, we conducted a computational analysis and discovered four short, conserved, linear motifs present in RNA-binding proteins. We show that mutation in one of the new motifs (WxHD) in an unstructured region of ALYREF reduced RNA binding and abolished the interaction with eIF4A3 and CBP80. Additionally, the mutation impaired proper localization to nuclear speckles and export of a spliced reporter mRNA. Our results reveal important details of the orchestrated recruitment of export factors during the formation of export competent mRNPs.


Subject(s)
Conserved Sequence , Nuclear Proteins/genetics , RNA Splicing , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics , Transcription Factors/genetics , Amino Acid Motifs , Biological Transport , Cloning, Molecular , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Exons , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Mutation , Nuclear Cap-Binding Protein Complex/genetics , Nuclear Cap-Binding Protein Complex/metabolism , Nuclear Proteins/metabolism , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism
14.
Blood ; 125(7): 1159-69, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25538044

ABSTRACT

The interaction between thrombopoietin (THPO) and its receptor c-Mpl regulates downstream cytokine signaling and platelet homeostasis. Hereditary mutations of c-Mpl can either result in loss-of-function and thrombocytopenia or in gain-of-function and thrombocythemia (HT), and are important models to analyze the mechanism of c-Mpl activity. We have analyzed the effect of the c-Mpl P106L gain-of-function and the nearby loss-of-function R102P and F104S mutations, which cause HT or thrombocytopenia, respectively, on posttranslational processing, intracellular trafficking, cell surface expression, and cell proliferation. In contrast to R102P and F104S, the P106L mutant confers cytokine-independent growth and stimulates downstream signaling after THPO treatment in Ba/F3 cells. Despite their opposite function, R102P and P106L, both lead to abnormal subcellular receptor distribution, lack of membrane localization, impaired glycosylation, and elevated THPO serum levels in effected patients. These findings indicate that the activation of downstream signaling by c-Mpl P106L does not require correct processing, trafficking, and cell surface expression of c-Mpl, whereas the negative feedback loop controlling THPO serum levels requires cell surface expression of the receptor. Thus, we propose that the P106L mutation functionally separates the activity of c-Mpl in downstream signaling from that in maintaining platelet homeostasis.


Subject(s)
Mutation, Missense , Receptors, Thrombopoietin/genetics , Thrombocytopenia/genetics , Thrombopoietin/metabolism , Adult , Amino Acid Substitution , Cells, Cultured , Child , Child, Preschool , Family , Female , HeLa Cells , Homeostasis/genetics , Humans , Leucine/genetics , Male , Pedigree , Proline/genetics , Receptors, Thrombopoietin/physiology , Signal Transduction/genetics , Thrombocytopenia/metabolism , Thrombocytosis/genetics , Young Adult
15.
Nucleic Acids Res ; 43(9): 4687-700, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25870412

ABSTRACT

In metazoan cells, spliced mRNAs are marked by the exon junction complex (EJC), a multi-protein complex that serves as a key regulator of post-transcriptional mRNA metabolism. Deposition of EJCs on mRNA is intimately linked to the splicing process. The spliceosomal protein CWC22 directly binds the core EJC-protein eIF4A3, guides it to the spliceosome and initiates EJC assembly. In addition, CWC22 is involved in the splicing process itself, but the molecular details of its dual function remain elusive. Here we analyze the mechanisms, by which CWC22 co-regulates pre-mRNA splicing and EJC assembly. We show that the core of CWC22 is sufficient to mediate both pre-mRNA splicing and EJC assembly. Nonetheless, both processes can be functionally uncoupled with an eIF4A3-binding deficient mutant of CWC22, which impedes EJC assembly. A C-terminal domain of CWC22 strongly enhances its spliceosomal interaction and likely regulates its function. High-throughput RNA-sequencing identifies global defects of pre-mRNA splicing and downregulation of diverse gene expression pathways in CWC22-depleted cells. We propose a model, in which CWC22 represents an integral component of the spliceosome and orchestrates pre-mRNA splicing and eIF4A3 binding to achieve global assembly of exon junction complexes.


Subject(s)
DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Exons , Nuclear Proteins/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Spliceosomes/metabolism , Binding Sites , Gene Expression , HEK293 Cells , Humans , Introns , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptidylprolyl Isomerase , Protein Binding , Protein Structure, Tertiary , RNA-Binding Proteins
16.
Pflugers Arch ; 468(6): 1013-28, 2016 06.
Article in English | MEDLINE | ID: mdl-27138169

ABSTRACT

Nonsense-mediated messenger RNA (mRNA) decay (NMD) is a quality control mechanism that degrades irregular or faulty mRNAs. NMD mainly degrades mRNAs, which contain a premature termination codon (PTC) and therefore encode a truncated protein. Furthermore, NMD alters the expression of different types of cellular mRNAs, the so-called endogenous NMD substrates. In this review, we focus on the impact of NMD on cellular and molecular physiology. We specify key classes of NMD substrates and provide a detailed overview of the physiological function of gene regulation by NMD. We also describe different mechanisms of NMD substrate degradation and how the regulation of the NMD machinery affects cellular physiology. Finally, we outline the physiological functions of central NMD factors.


Subject(s)
Nonsense Mediated mRNA Decay , Stress, Physiological/genetics , Animals , Humans , Immunity/genetics , MicroRNAs/genetics , Plants/genetics , Transcriptome
17.
RNA ; 20(10): 1579-92, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147240

ABSTRACT

Nonsense-mediated mRNA decay (NMD) eliminates different classes of mRNA substrates including transcripts with long 3' UTRs. Current models of NMD suggest that the long physical distance between the poly(A) tail and the termination codon reduces the interaction between cytoplasmic poly(A)-binding protein (PABPC1) and the eukaryotic release factor 3a (eRF3a) during translation termination. In the absence of PABPC1 binding, eRF3a recruits the NMD factor UPF1 to the terminating ribosome, triggering mRNA degradation. Here, we have used the MS2 tethering system to investigate the suppression of NMD by PABPC1. We show that tethering of PABPC1 between the termination codon and a long 3' UTR specifically inhibits NMD-mediated mRNA degradation. Contrary to the current model, tethered PABPC1 mutants unable to interact with eRF3a still efficiently suppress NMD. We find that the interaction of PABPC1 with eukaryotic initiation factor 4G (eIF4G), which mediates the circularization of mRNAs, is essential for NMD inhibition by tethered PABPC1. Furthermore, recruiting either eRF3a or eIF4G in proximity to an upstream termination codon antagonizes NMD. While tethering of an eRF3a mutant unable to interact with PABPC1 fails to suppress NMD, tethered eIF4G inhibits NMD in a PABPC1-independent manner, indicating a sequential arrangement of NMD antagonizing factors. In conclusion, our results establish a previously unrecognized link between translation termination, mRNA circularization, and NMD suppression, thereby suggesting a revised model for the activation of NMD at termination codons upstream of long 3' UTR.


Subject(s)
Codon, Nonsense/genetics , Eukaryotic Initiation Factor-4G/metabolism , Nonsense Mediated mRNA Decay/genetics , Peptide Termination Factors/metabolism , Poly(A)-Binding Protein I/metabolism , RNA, Messenger/metabolism , Blotting, Northern , Cytoplasm/metabolism , Eukaryotic Initiation Factor-4G/genetics , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Peptide Termination Factors/genetics , Poly A/metabolism , Poly(A)-Binding Protein I/genetics , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/metabolism
18.
Cell Mol Life Sci ; 72(23): 4523-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26283621

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is a translation-dependent, multistep process that degrades irregular or faulty messenger RNAs (mRNAs). NMD mainly targets mRNAs with a truncated open reading frame (ORF) due to premature termination codons (PTCs). In addition, NMD also regulates the expression of different types of endogenous mRNA substrates. A multitude of factors are involved in the tight regulation of the NMD mechanism. In this review, we focus on the molecular mechanism of mammalian NMD. Based on the published data, we discuss the involvement of translation termination in NMD initiation. Furthermore, we provide a detailed overview of the core NMD machinery, as well as several peripheral NMD factors, and discuss their function. Finally, we present an overview of diseases associated with NMD factor mutations and summarize the current state of treatment for genetic disorders caused by nonsense mutations.


Subject(s)
Genetic Predisposition to Disease , Nonsense Mediated mRNA Decay/physiology , 3' Untranslated Regions , Animals , Codon, Nonsense , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Humans , Mental Retardation, X-Linked/genetics , Mutation , Peptide Chain Termination, Translational/physiology , Phosphorylation , RNA Helicases , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Telomerase/genetics , Telomerase/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Nucleic Acids Res ; 42(4): 2673-86, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24271394

ABSTRACT

Nonsense-mediated decay (NMD) is a eukaryotic quality control pathway, involving conserved proteins UPF1, UPF2 and UPF3b, which detects and degrades mRNAs with premature stop codons. Human UPF2 comprises three tandem MIF4G domains and a C-terminal UPF1 binding region. MIF4G-3 binds UPF3b, but the specific functions of MIF4G-1 and MIF4G-2 are unknown. Crystal structures show that both MIF4G-1 and MIF4G-2 contain N-terminal capping helices essential for stabilization of the 10-helix MIF4G core and that MIF4G-2 interacts with MIF4G-3, forming a rigid assembly. The UPF2/UPF3b/SMG1 complex is thought to activate the kinase SMG1 to phosphorylate UPF1 in vivo. We identify MIF4G-3 as the binding site and in vitro substrate of SMG1 kinase and show that a ternary UPF2 MIF4G-3/UPF3b/SMG1 complex can form in vitro. Whereas in vivo complementation assays show that MIF4G-1 and MIF4G-2 are essential for NMD, tethering assays reveal that UPF2 truncated to only MIF4G-3 and the UPF1-binding region can still partially accomplish NMD. Thus UPF2 MIF4G-1 and MIF4G-2 appear to have a crucial scaffolding role, while MIF4G-3 is the key module required for triggering NMD.


Subject(s)
Nonsense Mediated mRNA Decay , Transcription Factors/chemistry , HeLa Cells , Humans , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
20.
Nucleic Acids Res ; 42(5): 3177-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24353314

ABSTRACT

NKAP is a highly conserved protein with roles in transcriptional repression, T-cell development, maturation and acquisition of functional competency and maintenance and survival of adult hematopoietic stem cells. Here we report the novel role of NKAP in splicing. With NKAP-specific antibodies we found that NKAP localizes to nuclear speckles. NKAP has an RS motif at the N-terminus followed by a highly basic domain and a DUF 926 domain at the C-terminal region. Deletion analysis showed that the basic domain is important for speckle localization. In pull-down experiments, we identified RNA-binding proteins, RNA helicases and splicing factors as interaction partners of NKAP, among them FUS/TLS. The FUS/TLS-NKAP interaction takes place through the RS domain of NKAP and the RGG1 and RGG3 domains of FUS/TLS. We analyzed the ability of NKAP to interact with RNA using in vitro splicing assays and found that NKAP bound both spliced messenger RNA (mRNA) and unspliced pre-mRNA. Genome-wide analysis using crosslinking and immunoprecipitation-seq revealed NKAP association with U1, U4 and U5 small nuclear RNA, and we also demonstrated that knockdown of NKAP led to an increase in pre-mRNA percentage. Our results reveal NKAP as nuclear speckle protein with roles in RNA splicing and processing.


Subject(s)
RNA/metabolism , Repressor Proteins/metabolism , Animals , Cell Nucleus , HEK293 Cells , HeLa Cells , Humans , Mice , Nuclear Proteins/analysis , Protein Structure, Tertiary , RNA Helicases/metabolism , RNA Splicing , RNA, Small Nuclear/metabolism , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/analysis , Repressor Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL