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











Publication year range
1.
Elife ; 122024 Oct 02.
Article in English | MEDLINE | ID: mdl-39356106

ABSTRACT

Previously, we showed that the germ cell-specific nuclear protein RBMXL2 represses cryptic splicing patterns during meiosis and is required for male fertility (Ehrmann et al., 2019). Here, we show that in somatic cells the similar yet ubiquitously expressed RBMX protein has similar functions. RBMX regulates a distinct class of exons that exceed the median human exon size. RBMX protein-RNA interactions are enriched within ultra-long exons, particularly within genes involved in genome stability, and repress the selection of cryptic splice sites that would compromise gene function. The RBMX gene is silenced during male meiosis due to sex chromosome inactivation. To test whether RBMXL2 might replace the function of RBMX during meiosis we induced expression of RBMXL2 and the more distantly related RBMY protein in somatic cells, finding each could rescue aberrant patterns of RNA processing caused by RBMX depletion. The C-terminal disordered domain of RBMXL2 is sufficient to rescue proper splicing control after RBMX depletion. Our data indicate that RBMX and RBMXL2 have parallel roles in somatic tissues and the germline that must have been conserved for at least 200 million years of mammalian evolution. We propose RBMX family proteins are particularly important for the splicing inclusion of some ultra-long exons with increased intrinsic susceptibility to cryptic splice site selection.


Subject(s)
Exons , RNA Splice Sites , RNA Splicing , RNA-Binding Proteins , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Humans , Exons/genetics , RNA Splice Sites/genetics , Male , Meiosis/genetics , Animals , Heterogeneous-Nuclear Ribonucleoproteins
2.
Nat Commun ; 15(1): 1964, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38467633

ABSTRACT

Despite the nuclear localization of the m6A machinery, the genomes of multiple exclusively-cytoplasmic RNA viruses, such as chikungunya (CHIKV) and dengue (DENV), are reported to be extensively m6A-modified. However, these findings are mostly based on m6A-Seq, an antibody-dependent technique with a high rate of false positives. Here, we address the presence of m6A in CHIKV and DENV RNAs. For this, we combine m6A-Seq and the antibody-independent SELECT and nanopore direct RNA sequencing techniques with functional, molecular, and mutagenesis studies. Following this comprehensive analysis, we find no evidence of m6A modification in CHIKV or DENV transcripts. Furthermore, depletion of key components of the host m6A machinery does not affect CHIKV or DENV infection. Moreover, CHIKV or DENV infection has no effect on the m6A machinery's localization. Our results challenge the prevailing notion that m6A modification is a general feature of cytoplasmic RNA viruses and underscore the importance of validating RNA modifications with orthogonal approaches.


Subject(s)
Adenosine/analogs & derivatives , Chikungunya Fever , Chikungunya virus , Dengue Virus , Dengue , Humans , Chikungunya virus/genetics , Dengue Virus/genetics , RNA, Viral/genetics , Antibodies, Viral
3.
Viruses ; 15(6)2023 06 16.
Article in English | MEDLINE | ID: mdl-37376680

ABSTRACT

The epitranscriptomic modification N6-methyladenosine (m6A) is a ubiquitous feature of the mammalian transcriptome. It modulates mRNA fate and dynamics to exert regulatory control over numerous cellular processes and disease pathways, including viral infection. Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation from the latent phase leads to the redistribution of m6A topology upon both viral and cellular mRNAs within infected cells. Here we investigate the role of m6A in cellular transcripts upregulated during KSHV lytic replication. Our results show that m6A is crucial for the stability of the GPRC5A mRNA, whose expression is induced by the KSHV latent-lytic switch master regulator, the replication and transcription activator (RTA) protein. Moreover, we demonstrate that GPRC5A is essential for efficient KSHV lytic replication by directly regulating NFκB signalling. Overall, this work highlights the central importance of m6A in modulating cellular gene expression to influence viral infection.


Subject(s)
Herpesvirus 8, Human , Herpesvirus 8, Human/genetics , Virus Latency/genetics , Cell Line, Tumor , Signal Transduction , RNA, Messenger/genetics , Virus Replication , Gene Expression Regulation, Viral
5.
Elife ; 82019 10 24.
Article in English | MEDLINE | ID: mdl-31647415

ABSTRACT

N6-methyladenosine (m6A) is the most abundant internal RNA modification of cellular mRNAs. m6A is recognised by YTH domain-containing proteins, which selectively bind to m6A-decorated RNAs regulating their turnover and translation. Using an m6A-modified hairpin present in the Kaposi's sarcoma associated herpesvirus (KSHV) ORF50 RNA, we identified seven members from the 'Royal family' as putative m6A readers, including SND1. RIP-seq and eCLIP analysis characterised the SND1 binding profile transcriptome-wide, revealing SND1 as an m6A reader. We further demonstrate that the m6A modification of the ORF50 RNA is critical for SND1 binding, which in turn stabilises the ORF50 transcript. Importantly, SND1 depletion leads to inhibition of KSHV early gene expression showing that SND1 is essential for KSHV lytic replication. This work demonstrates that members of the 'Royal family' have m6A-reading ability, greatly increasing their epigenetic functions beyond protein methylation.


Subject(s)
Adenosine/analogs & derivatives , Endonucleases/metabolism , Herpesvirus 8, Human/growth & development , Host-Pathogen Interactions , RNA, Messenger/metabolism , RNA, Viral/metabolism , Virus Replication , Adenosine/metabolism , Computational Biology , HEK293 Cells , Humans , Protein Binding , Sequence Analysis, RNA
6.
Mol Cell ; 75(2): 310-323.e8, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31104896

ABSTRACT

During gene expression, RNA export factors are mainly known for driving nucleo-cytoplasmic transport. While early studies suggested that the exon junction complex (EJC) provides a binding platform for them, subsequent work proposed that they are only recruited by the cap binding complex to the 5' end of RNAs, as part of TREX. Using iCLIP, we show that the export receptor Nxf1 and two TREX subunits, Alyref and Chtop, are recruited to the whole mRNA co-transcriptionally via splicing but before 3' end processing. Consequently, Alyref alters splicing decisions and Chtop regulates alternative polyadenylation. Alyref is recruited to the 5' end of RNAs by CBC, and our data reveal subsequent binding to RNAs near EJCs. We demonstrate that eIF4A3 stimulates Alyref deposition not only on spliced RNAs close to EJC sites but also on single-exon transcripts. Our study reveals mechanistic insights into the co-transcriptional recruitment of mRNA export factors and how this shapes the human transcriptome.


Subject(s)
DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Active Transport, Cell Nucleus/genetics , Binding Sites , DEAD-box RNA Helicases/chemistry , Eukaryotic Initiation Factor-4A/chemistry , Exons/genetics , Gene Expression Regulation/genetics , Humans , Nuclear Proteins/chemistry , Nucleocytoplasmic Transport Proteins/chemistry , Polyadenylation , RNA Transport/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , Transcription Factors/chemistry , Transcriptome/genetics
7.
Biochim Biophys Acta Gene Regul Mech ; 1862(3): 319-328, 2019 03.
Article in English | MEDLINE | ID: mdl-30290229

ABSTRACT

During synthesis, mRNA undergoes a number of modifications such as capping, splicing and polyadenylation. These processes are coupled with the orderly deposition of the TREX complex on the mRNA and subsequent recruitment of the NXF1-P15 heterodimer which stimulates the nuclear export of mature mRNAs. mRNAs also undergo a number of internal modifications, the most common of which is the N6­methyladenosine (m6A) modification. In this review we discuss the recent evidence of coupling between the m6A modification, RNA processing and export.


Subject(s)
Cell Nucleus/metabolism , Methyltransferases/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Adenine/analogs & derivatives , Adenine/metabolism , Animals , Exodeoxyribonucleases/metabolism , Humans , Phosphoproteins/metabolism , RNA, Messenger/genetics
8.
Sci Rep ; 8(1): 13827, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218090

ABSTRACT

N6-methyladenosine (m6A) is the most abundant internal modification of eukaryotic mRNA. This modification has previously been shown to alter the export kinetics for mRNAs though the molecular details surrounding this phenomenon remain poorly understood. Recruitment of the TREX mRNA export complex to mRNA is driven by transcription, 5' capping and pre-mRNA splicing. Here we identify a fourth mechanism in human cells driving the association of TREX with mRNA involving the m6A methylase complex. We show that the m6A complex recruits TREX to m6A modified mRNAs and this process is essential for their efficient export. TREX also stimulates recruitment of the m6A reader protein YTHDC1 to the mRNA and the m6A complex influences the interaction of TREX with YTHDC1. Together our studies reveal a key role for TREX in the export of m6A modified mRNAs.


Subject(s)
Adenosine/analogs & derivatives , Exodeoxyribonucleases/metabolism , Phosphoproteins/metabolism , RNA Transport/physiology , Active Transport, Cell Nucleus , Adenosine/metabolism , Adenosine/physiology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Exodeoxyribonucleases/physiology , HEK293 Cells , Humans , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/physiology , RNA Splicing/physiology , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
9.
Biochem J ; 473(19): 2911-35, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27679854

ABSTRACT

TRanscription and EXport (TREX) is a conserved multisubunit complex essential for embryogenesis, organogenesis and cellular differentiation throughout life. By linking transcription, mRNA processing and export together, it exerts a physiologically vital role in the gene expression pathway. In addition, this complex prevents DNA damage and regulates the cell cycle by ensuring optimal gene expression. As the extent of TREX activity in viral infections, amyotrophic lateral sclerosis and cancer emerges, the need for a greater understanding of TREX function becomes evident. A complete elucidation of the composition, function and interactions of the complex will provide the framework for understanding the molecular basis for a variety of diseases. This review details the known composition of TREX, how it is regulated and its cellular functions with an emphasis on mammalian systems.


Subject(s)
Gene Expression , Transcription Factors/physiology , Biological Transport , DNA Damage , Humans , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Nucleic Acids Res ; 43(4): 2353-66, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25662211

ABSTRACT

Cancer testis antigens (CTAs) represented a poorly characterized group of proteins whose expression is normally restricted to testis but are frequently up-regulated in cancer cells. Here we show that one CTA, Luzp4, is an mRNA export adaptor. It associates with the TREX mRNA export complex subunit Uap56 and harbours a Uap56 binding motif, conserved in other mRNA export adaptors. Luzp4 binds the principal mRNA export receptor Nxf1, enhances its RNA binding activity and complements Alyref knockdown in vivo. Whilst Luzp4 is up-regulated in a range of tumours, it appears preferentially expressed in melanoma cells where it is required for growth.


Subject(s)
Antigens, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Melanoma/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Antigens, Neoplasm/genetics , Cell Line , DNA-Binding Proteins/genetics , Humans , Melanoma/genetics , Nuclear Proteins/antagonists & inhibitors , Nucleocytoplasmic Transport Proteins/metabolism , RNA Transport , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
11.
RNA ; 21(3): 347-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605962

ABSTRACT

Splicing factor proline- and glutamine-rich (SFPQ) also commonly known as polypyrimidine tract-binding protein-associated-splicing factor (PSF) and its binding partner non-POU domain-containing octamer-binding protein (NONO/p54nrb), are highly abundant, multifunctional nuclear proteins. However, the exact role of this complex is yet to be determined. Following purification of the endogeneous SFPQ/NONO complex, mass spectrometry analysis identified a wide range of interacting proteins, including those involved in RNA processing, RNA splicing, and transcriptional regulation, consistent with a multifunctional role for SFPQ/NONO. In addition, we have identified several sites of arginine methylation in SFPQ/PSF using mass spectrometry and found that several arginines in the N-terminal domain of SFPQ/PSF are asymmetrically dimethylated. Furthermore, we find that the protein arginine N-methyltransferase, PRMT1, catalyzes this methylation in vitro and that this is antagonized by citrullination of SFPQ. Arginine methylation and citrullination of SFPQ/PSF does not affect complex formation with NONO. However, arginine methylation was shown to increase the association with mRNA in mRNP complexes in mammalian cells. Finally we show that the biochemical properties of the endogenous complex from cell lysates are significantly influenced by the ionic strength during purification. At low ionic strength, the SFPQ/NONO complex forms large heterogeneous protein assemblies or aggregates, preventing the purification of the SFPQ/NONO complex. The ability of the SFPQ/NONO complex to form varying protein assemblies, in conjunction with the effect of post-translational modifications of SFPQ modulating mRNA binding, suggests key roles affecting mRNP dynamics within the cell.


Subject(s)
Nuclear Matrix-Associated Proteins/genetics , Octamer Transcription Factors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic , Animals , Arginine/genetics , Arginine/metabolism , DNA-Binding Proteins , Gene Expression Regulation , HeLa Cells , Humans , Methylation , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nuclear Matrix-Associated Proteins/chemistry , Octamer Transcription Factors/chemistry , PTB-Associated Splicing Factor , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/genetics , RNA-Binding Proteins/chemistry , Repressor Proteins/genetics , Ribonucleoproteins/genetics
12.
Mol Microbiol ; 94(4): 828-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25231350

ABSTRACT

Candida albicans hyphae grow in a highly polarized fashion from their tips. This polarized growth requires the continuous delivery of secretory vesicles to the tip region. Vesicle delivery depends on Sec2p, the Guanine Exchange Factor (GEF) for the Rab GTPase Sec4p. GTP bound Sec4p is required for the transit of secretory vesicles from the trans-Golgi to sites of polarized growth. We previously showed that phosphorylation of Sec2p at residue S584 was necessary for Sec2p to support hyphal, but not yeast growth. Here we show that on secretory vesicles SEC2 mRNA is physically associated with Sec2p. Moreover, we show that the phosphorylation of S584 allows SEC2 mRNA to dissociate from Sec2p and we speculate that this is necessary for Sec2p function and/or translation. During hyphal extension, the growing tip may be separated from the nucleus by up to 15 µm. Transport of SEC2 mRNA on secretory vesicles to the tip localizes SEC2 translation to tip allowing a sufficient accumulation of this key protein at the site of polarized growth.


Subject(s)
Candida albicans/chemistry , Fungal Proteins/analysis , Hyphae/chemistry , RNA, Messenger/analysis , Secretory Vesicles/chemistry , Candida albicans/growth & development , Hyphae/growth & development , Phosphorylation , Protein Binding , Protein Processing, Post-Translational
13.
Brain ; 137(Pt 7): 2040-51, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24866055

ABSTRACT

GGGGCC repeat expansions of C9orf72 represent the most common genetic variant of amyotrophic lateral sclerosis and frontotemporal degeneration, but the mechanism of pathogenesis is unclear. Recent reports have suggested that the transcribed repeat might form toxic RNA foci that sequester various RNA processing proteins. Consensus as to the identity of the binding partners is missing and whole neuronal proteome investigation is needed. Using RNA fluorescence in situ hybridization we first identified nuclear and cytoplasmic RNA foci in peripheral and central nervous system biosamples from patients with amyotrophic lateral sclerosis with a repeat expansion of C9orf72 (C9orf72+), but not from those patients without a repeat expansion of C9orf72 (C9orf72-) or control subjects. Moreover, in the cases examined, the distribution of foci-positive neurons correlated with the clinical phenotype (t-test P < 0.05). As expected, RNA foci are ablated by RNase treatment. Interestingly, we identified foci in fibroblasts from an asymptomatic C9orf72+ carrier. We next performed pulldown assays, with GGGGCC5, in conjunction with mass spectrometry analysis, to identify candidate binding partners of the GGGGCC repeat expansion. Proteins containing RNA recognition motifs and involved in splicing, messenger RNA nuclear export and/or translation were significantly enriched. Immunohistochemistry in central nervous system tissue from C9orf72+ patients with amyotrophic lateral sclerosis demonstrated co-localization of RNA foci with SRSF2, hnRNP H1/F, ALYREF and hnRNP A1 in cerebellar granule cells and with SRSF2, hnRNP H1/F and ALYREF in motor neurons, the primary target of pathology in amyotrophic lateral sclerosis. Direct binding of proteins to GGGGCC repeat RNA was confirmed in vitro by ultraviolet-crosslinking assays. Co-localization was only detected in a small proportion of RNA foci, suggesting dynamic sequestration rather than irreversible binding. Additional immunohistochemistry demonstrated that neurons with and without RNA foci were equally likely to show nuclear depletion of TDP-43 (χ(2) P = 0.75) or poly-GA dipeptide repeat protein inclusions (χ(2) P = 0.46). Our findings suggest two non-exclusive pathogenic mechanisms: (i) functional depletion of RNA-processing proteins resulting in disruption of messenger RNA splicing; and (ii) licensing of expanded C9orf72 pre-messenger RNA for nuclear export by inappropriate association with messenger RNA export adaptor protein(s) leading to cytoplasmic repeat associated non-ATG translation and formation of potentially toxic dipeptide repeat protein.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Repeat Expansion/genetics , Proteins/genetics , RNA-Binding Proteins/metabolism , Adenosine Triphosphate/pharmacokinetics , Amyotrophic Lateral Sclerosis/pathology , Biotinylation , Brain/pathology , C9orf72 Protein , Female , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Humans , Male , Mass Spectrometry , Neurons/pathology , Nuclear Proteins/metabolism , Phosphorus Isotopes/pharmacokinetics , Protein Binding/drug effects , RNA-Binding Proteins/genetics , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors , Transcription Factors/metabolism
14.
PLoS Pathog ; 10(2): e1003907, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550725

ABSTRACT

The essential herpesvirus adaptor protein HVS ORF57, which has homologs in all other herpesviruses, promotes viral mRNA export by utilizing the cellular mRNA export machinery. ORF57 protein specifically recognizes viral mRNA transcripts, and binds to proteins of the cellular transcription-export (TREX) complex, in particular ALYREF. This interaction introduces viral mRNA to the NXF1 pathway, subsequently directing it to the nuclear pore for export to the cytoplasm. Here we have used a range of techniques to reveal the sites for direct contact between RNA and ORF57 in the absence and presence of ALYREF. A binding site within ORF57 was characterized which recognizes specific viral mRNA motifs. When ALYREF is present, part of this ORF57 RNA binding site, composed of an α-helix, binds preferentially to ALYREF. This competitively displaces viral RNA from the α-helix, but contact with RNA is still maintained by a flanking region. At the same time, the flexible N-terminal domain of ALYREF comes into contact with the viral RNA, which becomes engaged in an extensive network of synergistic interactions with both ALYREF and ORF57. Transfer of RNA to ALYREF in the ternary complex, and involvement of individual ORF57 residues in RNA recognition, were confirmed by UV cross-linking and mutagenesis. The atomic-resolution structure of the ORF57-ALYREF interface was determined, which noticeably differed from the homologous ICP27-ALYREF structure. Together, the data provides the first site-specific description of how viral mRNA is locked by a herpes viral adaptor protein in complex with cellular ALYREF, giving herpesvirus access to the cellular mRNA export machinery. The NMR strategy used may be more generally applicable to the study of fuzzy protein-protein-RNA complexes which involve flexible polypeptide regions.


Subject(s)
Herpesviridae Infections/metabolism , Host-Parasite Interactions/physiology , Nuclear Proteins/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Virus Infections/metabolism , Active Transport, Cell Nucleus/physiology , Herpesvirus 2, Saimiriine/chemistry , Herpesvirus 2, Saimiriine/metabolism , Herpesvirus 2, Saimiriine/pathogenicity , Humans , Nuclear Proteins/chemistry , Protein Structure, Quaternary , RNA Transport/physiology , RNA, Viral/analysis , RNA-Binding Proteins/chemistry , Repressor Proteins/chemistry , Trans-Activators/chemistry , Transcription Factors/chemistry
15.
PLoS One ; 8(6): e67676, 2013.
Article in English | MEDLINE | ID: mdl-23826332

ABSTRACT

The TREX complex couples nuclear mRNA processing events with subsequent export to the cytoplasm. TREX also acts as a binding platform for the mRNA export receptor Nxf1. The sites of mRNA transcription and processing within the nucleus have been studied extensively. However, little is known about where TREX assembly takes place and where Nxf1 is recruited to TREX to form the export competent mRNP. Here we have used sensitized emission Förster resonance energy transfer (FRET) and fluorescence lifetime imaging (FLIM)-FRET, to produce a spatial map in living cells of the sites for the interaction of two TREX subunits, Alyref and Chtop, with Nxf1. Prominent assembly sites for export factors are found in the vicinity of nuclear speckles in regions known to be involved in transcription, splicing and exon junction complex formation highlighting the close coupling of mRNA export with mRNP biogenesis.


Subject(s)
Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Interaction Mapping , RNA Transport , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Cell Nucleus/metabolism , Cell Survival , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Protein Binding , Protein Transport , Transcription, Genetic
16.
EMBO J ; 32(3): 473-86, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23299939

ABSTRACT

The TREX complex couples nuclear pre-mRNA processing with mRNA export and contains multiple protein components, including Uap56, Alyref, Cip29 and the multi-subunit THO complex. Here, we have identified Chtop as a novel TREX component. We show that both Chtop and Alyref activate the ATPase and RNA helicase activities of Uap56 and that Uap56 functions to recruit both Alyref and Chtop onto mRNA. As observed with the THO complex subunit Thoc5, Chtop binds to the NTF2-like domain of Nxf1, and this interaction requires arginine methylation of Chtop. Using RNAi, we show that co-knockdown of Alyref and Chtop results in a potent mRNA export block. Chtop binds to Uap56 in a mutually exclusive manner with Alyref, and Chtop binds to Nxf1 in a mutually exclusive manner with Thoc5. However, Chtop, Thoc5 and Nxf1 exist in a single complex in vivo. Together, our data indicate that TREX and Nxf1 undergo dynamic remodelling, driven by the ATPase cycle of Uap56 and post-translational modifications of Chtop.


Subject(s)
Exodeoxyribonucleases/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Transcription Factors/metabolism , Blotting, Western , DEAD-box RNA Helicases/metabolism , DNA Methylation , DNA Primers/genetics , Genetic Complementation Test , Humans , In Situ Hybridization, Fluorescence , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , RNA Interference , RNA-Binding Proteins/metabolism , Transcription Factors/genetics
17.
Nat Commun ; 3: 1006, 2012.
Article in English | MEDLINE | ID: mdl-22893130

ABSTRACT

The metazoan TREX complex is recruited to mRNA during nuclear RNA processing and functions in exporting mRNA to the cytoplasm. Nxf1 is an mRNA export receptor, which binds processed mRNA and transports it through the nuclear pore complex. At present, the relationship between TREX and Nxf1 is not understood. Here we show that Nxf1 uses an intramolecular interaction to inhibit its own RNA-binding activity. When the TREX subunits Aly and Thoc5 make contact with Nxf1, Nxf1 is driven into an open conformation, exposing its RNA-binding domain, allowing RNA binding. Moreover, the combined knockdown of Aly and Thoc5 markedly reduces the amount of Nxf1 bound to mRNA in vivo and also causes a severe mRNA export block. Together, our data indicate that TREX provides a license for mRNA export by driving Nxf1 into a conformation capable of binding mRNA.


Subject(s)
Exodeoxyribonucleases/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , RNA Transport , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Biochem Soc Trans ; 40(4): 842-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22817745

ABSTRACT

Melioidosis is a disease caused by infection with Burkholderia pseudomallei. The molecular basis for the pathogenicity of B. pseudomallei is poorly understood. However, recent work has identified the first toxin from this bacterium and shown that it inhibits host protein synthesis. Here, we review the illness that is potentially associated with biological warfare, the pathogen and its deadly molecular mechanism of action, as well as therapeutic developments that may follow.


Subject(s)
Bacterial Toxins/metabolism , Burkholderia pseudomallei/pathogenicity , Melioidosis/metabolism , Animals , Burkholderia pseudomallei/metabolism , Humans , Protein Biosynthesis
19.
Nat Neurosci ; 15(7): 962-9, 2012 Jun 17.
Article in English | MEDLINE | ID: mdl-22706270

ABSTRACT

Temporal regulation of embryonic neurogenesis is controlled by hypostable transcription factors. The mechanism of the process is unclear. Here we show that the RNase III Drosha and DGCR8 (also known as Pasha), key components of the microRNA (miRNA) microprocessor, have important functions in mouse neurogenesis. Loss of microprocessor in forebrain neural progenitors resulted in a loss of stem cell character and precocious differentiation whereas Dicer deficiency did not. Drosha negatively regulated expression of the transcription factors Neurogenin 2 (Ngn2) and NeuroD1 whereas forced Ngn2 expression phenocopied the loss of Drosha. Neurog2 mRNA contains evolutionarily conserved hairpins with similarities to pri-miRNAs, and associates with the microprocessor in neural progenitors. We uncovered a Drosha-dependent destabilization of Neurog2 mRNAs consistent with microprocessor cleavage at hairpins. Our findings implicate direct and miRNA-independent destabilization of proneural mRNAs by the microprocessor, which facilitates neural stem cell (NSC) maintenance by blocking accumulation of differentiation and determination factors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Gene Expression Regulation , MicroRNAs , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Ribonuclease III/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Pregnancy , Ribonuclease III/antagonists & inhibitors
20.
Nucleic Acids Res ; 40(7): 3232-44, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22140111

ABSTRACT

SRSF2 is a prototypical SR protein which plays important roles in the alternative splicing of pre-mRNA. It has been shown to be involved in regulatory pathways for maintaining genomic stability and play important roles in regulating key receptors in the heart. We report here the solution structure of the RNA recognition motifs (RRM) domain of free human SRSF2 (residues 9-101). Compared with other members of the SR protein family, SRSF2 structure has a longer L3 loop region. The conserved aromatic residue in the RNP2 motif is absent in SRSF2. Calorimetric titration shows that the RNA sequence 5'AGCAGAGUA3' binds SRSF2 with a K(d) of 61 ± 1 nM and a 1:1 stoichiometry. NMR and mutagenesis experiments reveal that for SFSF2, the canonical ß1 and ß3 interactions are themselves not sufficient for effective RNA binding; the additional loop L3 is crucial for RNA complex formation. A comparison is made between the structures of SRSF2-RNA complex with other known RNA complexes of SR proteins. We conclude that interactions involving the L3 loop, N- and C-termini of the RRM domain are collectively important for determining selectivity between the protein and RNA.


Subject(s)
Nuclear Proteins/chemistry , RNA/chemistry , Ribonucleoproteins/chemistry , Amino Acid Sequence , Binding Sites , Calorimetry , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , RNA/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sequence Homology, Amino Acid , Serine-Arginine Splicing Factors
SELECTION OF CITATIONS
SEARCH DETAIL