Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Annu Rev Microbiol ; 74: 387-407, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32660384

ABSTRACT

Ro60 ribonucleoproteins (RNPs), composed of the ring-shaped Ro 60-kDa (Ro60) protein and noncoding RNAs called Y RNAs, are present in all three domains of life. Ro60 was first described as an autoantigen in patients with rheumatic disease, and Ro60 orthologs have been identified in 3% to 5% of bacterial genomes, spanning the majority of phyla. Their functions have been characterized primarily in Deinococcus radiodurans, the first sequenced bacterium with a recognizable ortholog. In D. radiodurans, the Ro60 ortholog enhances the ability of 3'-to-5' exoribonucleases to degrade structured RNA during several forms of environmental stress. Y RNAs are regulators that inhibit or allow the interactions of Ro60 with other proteins and RNAs. Studies of Ro60 RNPs in other bacteria hint at additional functions, since the most conserved Y RNA contains a domain that is a close tRNA mimic and Ro60 RNPs are often encoded adjacent to components of RNA repair systems.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Untranslated/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Bacteria/chemistry , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Nucleic Acid Conformation , RNA Stability , Ribonucleoproteins/classification
2.
Nucleic Acids Res ; 48(12): 6919-6930, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32469055

ABSTRACT

Noncoding Y RNAs are abundant in animal cells and present in many bacteria. These RNAs are bound and stabilized by Ro60, a ring-shaped protein that is a target of autoantibodies in patients with systemic lupus erythematosus. Studies in bacteria revealed that Y RNA tethers Ro60 to a ring-shaped exoribonuclease, forming a double-ringed RNP machine specialized for structured RNA degradation. In addition to functioning as a tether, the bacterial RNA gates access of substrates to the Ro60 cavity. To identify roles for Y RNAs in mammals, we used CRISPR to generate mouse embryonic stem cells lacking one or both of the two murine Y RNAs. Despite reports that animal cell Y RNAs are essential for DNA replication, cells lacking these RNAs divide normally. However, Ro60 levels are reduced, revealing that Y RNA binding is required for Ro60 to accumulate to wild-type levels. Y RNAs regulate the subcellular location of Ro60, since Ro60 is reduced in the cytoplasm and increased in nucleoli when Y RNAs are absent. Last, we show that Y RNAs tether Ro60 to diverse effector proteins to generate specialized RNPs. Together, our data demonstrate that the roles of Y RNAs are intimately connected to that of their Ro60 partner.


Subject(s)
Autoantigens/genetics , RNA, Small Cytoplasmic/genetics , RNA, Untranslated/genetics , Ribonucleoproteins/genetics , Animals , Autoantibodies/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cytoplasm/genetics , Humans , Mice , Nucleic Acid Conformation , RNA Stability/genetics , RNA, Untranslated/ultrastructure
3.
J Cell Biol ; 218(8): 2564-2582, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31308215

ABSTRACT

A defining feature of embryonic stem cells (ESCs) is the ability to differentiate into all three germ layers. Pluripotency is maintained in part by a unique transcription network that maintains expression of pluripotency-specific transcription factors and represses developmental genes. While the mechanisms that establish this transcription network are well studied, little is known of the posttranscriptional surveillance pathways that degrade differentiation-related RNAs. We report that the surveillance pathway mediated by the RNA exosome nuclease complex represses ESC differentiation. Depletion of the exosome expedites differentiation of human ESCs into all three germ layers. LINE-1 retrotransposons and specific miRNAs, lncRNAs, and mRNAs that encode developmental regulators or affect their expression are all bound by the exosome and increase in level upon exosome depletion. The exosome restrains differentiation in part by degrading transcripts encoding FOXH1, a transcription factor crucial for mesendoderm formation. Our studies establish the exosome as a regulator of human ESC differentiation and reveal the importance of RNA decay in maintaining pluripotency.


Subject(s)
Cell Differentiation , Exosome Multienzyme Ribonuclease Complex/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Cross-Linking Reagents/chemistry , Endoderm/embryology , Endoderm/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Long Interspersed Nucleotide Elements/genetics , Mesoderm/embryology , Mesoderm/metabolism , MicroRNAs/genetics , Phenotype , RNA/isolation & purification , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic , Transgenes
4.
Microbiol Spectr ; 6(4)2018 07.
Article in English | MEDLINE | ID: mdl-30006996

ABSTRACT

Y RNAs are noncoding RNAs (ncRNAs) that are present in most animal cells and also in many bacteria. These RNAs were discovered because they are bound by the Ro60 protein, a major target of autoantibodies in patients with some systemic autoimmune rheumatic diseases. Studies of Ro60 and Y RNAs in Deinococcus radiodurans, the first sequenced bacterium with a Ro60 ortholog, revealed that they function with 3'-to-5' exoribonucleases to alter the composition of RNA populations during some forms of environmental stress. In the best-characterized example, Y RNA tethers the Ro60 protein to the exoribonuclease polynucleotide phosphorylase, allowing this exoribonuclease to degrade structured RNAs more effectively. Y RNAs can also function as gates to regulate access of other RNAs to the Ro60 central cavity. Recent studies in the enteric bacterium Salmonella enterica serovar Typhimurium resulted in the discovery that Y RNAs are widely present in bacteria. Remarkably, the most-conserved subclass of bacterial Y RNAs contains a domain that mimics tRNA. In this review, we discuss the structure, conservation, and known functions of bacterial Y RNAs as well as the certainty that more bacterial Y RNAs and additional roles for these ncRNAs remain to be uncovered.


Subject(s)
RNA, Bacterial/metabolism , RNA, Transfer/metabolism , RNA, Untranslated/metabolism , Animals , Autoantigens , Bacteria/classification , Bacteria/genetics , Base Sequence , Binding Sites , Deinococcus , Exoribonucleases , Humans , RNA, Bacterial/genetics , RNA, Small Cytoplasmic , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins , Salmonella enterica , Stress, Psychological
5.
Sci Transl Med ; 10(434)2018 03 28.
Article in English | MEDLINE | ID: mdl-29593104

ABSTRACT

The earliest autoantibodies in lupus are directed against the RNA binding autoantigen Ro60, but the triggers against this evolutionarily conserved antigen remain elusive. We identified Ro60 orthologs in a subset of human skin, oral, and gut commensal bacterial species and confirmed the presence of these orthologs in patients with lupus and healthy controls. Thus, we hypothesized that commensal Ro60 orthologs may trigger autoimmunity via cross-reactivity in genetically susceptible individuals. Sera from human anti-Ro60-positive lupus patients immunoprecipitated commensal Ro60 ribonucleoproteins. Human Ro60 autoantigen-specific CD4 memory T cell clones from lupus patients were activated by skin and mucosal Ro60-containing bacteria, supporting T cell cross-reactivity in humans. Further, germ-free mice spontaneously initiated anti-human Ro60 T and B cell responses and developed glomerular immune complex deposits after monocolonization with a Ro60 ortholog-containing gut commensal, linking anti-Ro60 commensal responses in vivo with the production of human Ro60 autoantibodies and signs of autoimmunity. Together, these data support that colonization with autoantigen ortholog-producing commensal species may initiate and sustain chronic autoimmunity in genetically predisposed individuals. The concept of commensal ortholog cross-reactivity may apply more broadly to autoimmune diseases and lead to novel treatment approaches aimed at defined commensal species.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Lupus Nephritis/immunology , Ribonucleoproteins/immunology , Animals , Autoantigens/chemistry , Autoantigens/genetics , Cell Proliferation , Female , Humans , Male , Mice , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , T-Lymphocytes/metabolism
6.
Chem Rev ; 118(8): 4422-4447, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29023106

ABSTRACT

Numerous surveillance pathways sculpt eukaryotic transcriptomes by degrading unneeded, defective, and potentially harmful noncoding RNAs (ncRNAs). Because aberrant and excess ncRNAs are largely degraded by exoribonucleases, a key characteristic of these RNAs is an accessible, protein-free 5' or 3' end. Most exoribonucleases function with cofactors that recognize ncRNAs with accessible 5' or 3' ends and/or increase the availability of these ends. Noncoding RNA surveillance pathways were first described in budding yeast, and there are now high-resolution structures of many components of the yeast pathways and significant mechanistic understanding as to how they function. Studies in human cells are revealing the ways in which these pathways both resemble and differ from their yeast counterparts, and are also uncovering numerous pathways that lack equivalents in budding yeast. In this review, we describe both the well-studied pathways uncovered in yeast and the new concepts that are emerging from studies in mammalian cells. We also discuss the ways in which surveillance pathways compete with chaperone proteins that transiently protect nascent ncRNA ends from exoribonucleases, with partner proteins that sequester these ends within RNPs, and with end modification pathways that protect the ends of some ncRNAs from nucleases.


Subject(s)
DNA, Fungal/genetics , RNA, Untranslated/genetics , Saccharomyces cerevisiae/genetics , Animals , Exosomes/metabolism , Humans
7.
Genes Dev ; 29(6): 646-57, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25792599

ABSTRACT

Although all retroviruses recruit host cell RNAs into virions, both the spectrum of RNAs encapsidated and the mechanisms by which they are recruited remain largely unknown. Here, we used high-throughput sequencing to obtain a comprehensive description of the RNAs packaged by a model retrovirus, murine leukemia virus. The major encapsidated host RNAs are noncoding RNAs (ncRNAs) and members of the VL30 class of endogenous retroviruses. Remarkably, although Moloney leukemia virus (MLV) assembles in the cytoplasm, precursors to specific tRNAs, small nuclear RNAs (snRNAs), and small nucleolar RNAs (snoRNAs) are all enriched in virions. Consistent with their cytoplasmic recruitment, packaging of both pre-tRNAs and U6 snRNA requires the nuclear export receptor Exportin-5. Adenylated and uridylated forms of these RNAs accumulate in cells and virions when the cytoplasmic exoribonuclease DIS3L2 and subunits of the RNA exosome are depleted. Together, our data reveal that MLV recruits RNAs from a novel host cell surveillance pathway in which unprocessed and unneeded nuclear ncRNAs are exported to the cytoplasm for degradation.


Subject(s)
Host-Pathogen Interactions/physiology , Moloney murine leukemia virus/metabolism , RNA, Untranslated/metabolism , Animals , Cytoplasm , Gene Expression Profiling , Mice , NIH 3T3 Cells , Protein Transport , Proteolysis , Vault Ribonucleoprotein Particles/metabolism , Virion/metabolism
8.
RNA ; 20(11): 1715-24, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25232022

ABSTRACT

Many bacteria encode an ortholog of the Ro60 autoantigen, a ring-shaped protein that is bound in animal cells to noncoding RNAs (ncRNAs) called Y RNAs. Studies in Deinococcus radiodurans revealed that Y RNA tethers Ro60 to polynucleotide phosphorylase, specializing this exoribonuclease for structured RNA degradation. Although Ro60 orthologs are present in a wide range of bacteria, Y RNAs have been detected in only two species, making it unclear whether these ncRNAs are common Ro60 partners in bacteria. In this study, we report that likely Y RNAs are encoded near Ro60 in >250 bacterial and phage species. By comparing conserved features, we discovered that at least one Y RNA in each species contains a domain resembling tRNA. We show that these RNAs contain nucleotide modifications characteristic of tRNA and are substrates for several enzymes that recognize tRNAs. Our studies confirm the importance of Y RNAs in bacterial physiology and identify a new class of ncRNAs that mimic tRNA.


Subject(s)
Bacteria/genetics , RNA, Bacterial/isolation & purification , RNA, Bacterial/metabolism , RNA, Untranslated/isolation & purification , RNA, Untranslated/metabolism , Animals , Bacteria/classification , Databases, Genetic , Humans , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Transfer/metabolism , RNA, Untranslated/chemistry , Ribonucleoproteins/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid
9.
RNA Biol ; 10(10): 1602-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24036917

ABSTRACT

Non-coding RNAs (ncRNAs) called Y RNAs are abundant components of both animal cells and a variety of bacteria. In all species examined, these ~100 nt RNAs are bound to the Ro 60 kDa (Ro60) autoantigen, a ring-shaped protein that also binds misfolded ncRNAs in some vertebrate nuclei. Although the function of Ro60 RNPs has been mysterious, we recently reported that a bacterial Y RNA tethers Ro60 to the 3' to 5' exoribonuclease polynucleotide phosphorylase (PNPase) to form RYPER (Ro60/Y RNA/PNPase Exoribonuclease RNP), a new RNA degradation machine. PNPase is a homotrimeric ring that degrades single-stranded RNA, and Y RNA-mediated tethering of Ro60 increases the effectiveness of PNPase in degrading structured RNAs. Single particle electron microscopy of RYPER suggests that RNA threads through the Ro60 ring into the PNPase cavity. Further studies indicate that Y RNAs may also act as gates to regulate entry of RNA substrates into the Ro60 channel. These findings reveal novel functions for Y RNAs and raise questions about how the bacterial findings relate to the roles of these ncRNAs in animal cells. Here we review the literature on Y RNAs, highlighting their close relationship with Ro60 proteins and the hypothesis that these ncRNAs function generally to tether Ro60 rings to diverse RNA-binding proteins.


Subject(s)
RNA, Untranslated/metabolism , Amino Acid Sequence , Animals , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Untranslated/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism
10.
J Immunol ; 191(1): 110-6, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23698747

ABSTRACT

Cardiac neonatal lupus (NL) is presumed to arise from maternal autoantibody targeting an intracellular ribonucleoprotein, Ro60, which binds noncoding Y RNA and only becomes accessible to autoantibodies during apoptosis. Despite the importance of Ro60 trafficking in the development of cardiac NL, the mechanism underlying cell surface exposure is unknown. To evaluate the influence of Y RNA on the subcellular location of Ro60 during apoptosis and activation of macrophages, stable Ro60 knockout murine fibroblasts expressing wild-type or mutated FLAG-Ro60 were assessed. FLAG3-Ro60(K170A R174A) binds Y RNA, whereas FLAG3-Ro60(H187S) does not bind Y RNA; fibroblasts expressing these constructs showed equivalent intracellular expression of Ro60. In contrast, apoptotic fibroblasts containing FLAG3-Ro60(K170A R174A) were bound by anti-Ro60, whereas FLAG3-Ro60(H187S) was not surface expressed. RNA interference of mY3 RNA in wild-type fibroblasts inhibited surface translocation of Ro60 during apoptosis, whereas depletion of mY1 RNA did not affect Ro60 exposure. Furthermore, Ro60 was not exposed following overexpression of mY1 in the mY3-depleted fibroblasts. In an in vitro model of anti-Ro60-mediated injury, Y RNA was shown to be an obligate factor for TLR-dependent activation of macrophages challenged with anti-Ro60-opsonized apoptotic fibroblasts. Murine Y3 RNA is a necessary factor to support the surface translocation of Ro60, which is pivotal to the formation of immune complexes on apoptotic cells and a TLR-dependent proinflammatory cascade. Accordingly, the Y3 RNA moiety of the Ro60 ribonucleoprotein imparts a critical role in the pathogenicity of maternal anti-Ro60 autoantibodies.


Subject(s)
Heart Diseases/immunology , Heart Diseases/metabolism , Lupus Erythematosus, Systemic/metabolism , RNA, Untranslated/metabolism , Ribonucleoproteins/metabolism , Adult , Animals , Animals, Newborn , Autoantibodies/metabolism , Cells, Cultured , Child , Coculture Techniques , Heart Diseases/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/immunology , Subcellular Fractions/immunology , Surface Properties
11.
Trends Genet ; 28(7): 306-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22475369

ABSTRACT

Nuclear noncoding RNA (ncRNA) surveillance pathways play key roles in shaping the steady-state transcriptomes of eukaryotic cells. Defective and unneeded ncRNAs are primarily degraded by exoribonucleases that rely on protein cofactors to identify these RNAs. Recent studies have begun to elucidate both the mechanisms by which these cofactors recognize aberrant RNAs and the features that mark RNAs for degradation. One crucial RNA determinant is the presence of an accessible end; in addition, the failure of aberrant RNAs to fold into compact structures and assemble with specific binding proteins probably also contributes to their recognition and subsequent degradation. To date, ncRNA surveillance has been most extensively studied in budding yeast. However, mammalian cells possess nucleases and cofactors that have no known yeast counterparts, indicating that RNA surveillance pathways may be more complex in metazoans. Importantly, there is evidence that the failure of ncRNA surveillance pathways contributes to human disease.


Subject(s)
Cell Nucleus/metabolism , RNA, Nuclear/metabolism , RNA, Untranslated/metabolism , Exosomes/metabolism , Humans , RNA Folding , RNA Stability , RNA, Nuclear/chemistry , RNA, Untranslated/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptome
12.
RNA ; 18(1): 100-10, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114317

ABSTRACT

The Ro 60-kDa autoantigen, a ring-shaped RNA-binding protein, traffics between the nucleus and cytoplasm in vertebrate cells. In some vertebrate nuclei, Ro binds misfolded noncoding RNAs and may function in quality control. In the cytoplasm, Ro binds noncoding RNAs called Y RNAs. Y RNA binding blocks a nuclear accumulation signal, retaining Ro in the cytoplasm. Following UV irradiation, this signal becomes accessible, allowing Ro to accumulate in nuclei. To investigate how other cellular components influence the function and subcellular location of Ro, we identified several proteins that copurify with the mouse Ro protein. Here, we report that the zipcode-binding protein ZBP1 influences the subcellular localization of both Ro and the Y3 RNA. Binding of ZBP1 to the Ro/Y3 complex increases after UV irradiation and requires the Y3 RNA. Despite the lack of an identifiable CRM1-dependent export signal, nuclear export of Ro is sensitive to the CRM1 inhibitor leptomycin B. In agreement with a previous report, we find that ZBP1 export is partly dependent on CRM1. Both Ro and Y3 RNA accumulate in nuclei when ZBP1 is depleted. Our data indicate that ZBP1 may function as an adapter to export the Ro/Y3 RNA complex from nuclei.


Subject(s)
Autoantigens/metabolism , Cell Nucleus/metabolism , Glycoproteins/metabolism , RNA, Small Cytoplasmic/metabolism , RNA, Untranslated/metabolism , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Fatty Acids, Unsaturated/pharmacology , Glycoproteins/genetics , Karyopherins/antagonists & inhibitors , Karyopherins/metabolism , Mice , RNA-Binding Proteins , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Ultraviolet Rays , Exportin 1 Protein
13.
Wiley Interdiscip Rev RNA ; 2(5): 686-99, 2011.
Article in English | MEDLINE | ID: mdl-21823229

ABSTRACT

All cells contain an enormous variety of ribonucleoprotein (RNP) complexes that function in diverse processes. Although the mechanisms by which many of these RNPs contribute to cell metabolism are well understood, the roles of others are only now beginning to be revealed. A member of this latter category, the Ro 60-kDa protein and its associated noncoding Y RNAs, was discovered because the protein component is a frequent target of the autoimmune response in patients with the rheumatic diseases systemic lupus erythematosus and Sjögren's syndrome. Recent studies have shown that Ro is ring shaped, binds the single-stranded ends of misfolded noncoding RNAs in its central cavity, and may function in noncoding RNA quality control. Although Ro is not present in yeast, many bacterial genomes contain potential Ro orthologs. In the radiation-resistant eubacterium Deinococcus radiodurans, the Ro ortholog functions with exoribonucleases during stress-induced changes in RNA metabolism. Moreover, in both D. radiodurans and animal cells, Ro is involved in the response to multiple types of environmental stress. Finally, Y RNAs can influence the subcellular location of Ro, inhibit access of the central cavity to other RNAs, and may also act as binding sites for proteins that influence Ro function. WIREs RNA 2011 2 686-699 DOI: 10.1002/wrna.85 For further resources related to this article, please visit the WIREs website.


Subject(s)
RNA, Untranslated/metabolism , Ribonucleoproteins/metabolism , Animals , Autoantigens/chemistry , Autoantigens/genetics , Autoantigens/metabolism , Deinococcus/genetics , Deinococcus/metabolism , Embryonic Stem Cells/metabolism , Female , Humans , Mice , Models, Molecular , Nucleic Acid Conformation , Oocytes/metabolism , Phylogeny , RNA Stability , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal, 5S/metabolism , RNA, Small Cytoplasmic/chemistry , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , RNA, Small Nuclear/metabolism , RNA, Untranslated/genetics , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Stress, Physiological , Xenopus laevis
14.
J Virol ; 83(23): 12526-34, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776129

ABSTRACT

Moloney murine leukemia virus (MLV) selectively encapsidates host mY1 and mY3 RNAs. These noncoding RNA polymerase III transcripts are normally complexed with the Ro60 and La proteins, which are autoantigens associated with rheumatic disease that function in RNA biogenesis and quality control. Here, MLV replication and mY RNA packaging were analyzed using Ro60 knockout embryonic fibroblasts, which contain only approximately 3% as much mY RNA as wild-type cells. Virus spread at the same rate in wild-type and Ro knockout cells. Surprisingly, MLV virions shed by Ro60 knockout cells continued to package high levels of mY1 and mY3 (about two copies of each) like those from wild-type cells, even though mY RNAs were barely detectable within producer cells. As a result, for MLV produced in Ro60 knockout cells, encapsidation selectivity from among all cell RNAs was even higher for mY RNAs than for the viral genome. Whereas mY RNAs are largely cytoplasmic in wild-type cells, fractionation of knockout cells revealed that the residual mY RNAs were relatively abundant in nuclei, likely reflecting the fact that most mY RNAs were degraded shortly after transcription in the absence of Ro60. Together, these data suggest that these small, labile host RNAs may be recruited at a very early stage of their biogenesis and may indicate an intersection of retroviral assembly and RNA quality control pathways.


Subject(s)
Moloney murine leukemia virus/physiology , RNA, Small Cytoplasmic/metabolism , Virus Assembly , Animals , Autoantigens , Cell Line , Fibroblasts/virology , Mice , Mice, Knockout , Ribonucleoproteins/deficiency
15.
Mol Biol Cell ; 20(5): 1555-64, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19116308

ABSTRACT

The Ro autoantigen is a ring-shaped RNA-binding protein that binds misfolded RNAs in nuclei and is proposed to function in quality control. In the cytoplasm, Ro binds noncoding RNAs, called Y RNAs, that inhibit access of Ro to other RNAs. Ro also assists survival of mammalian cells and at least one bacterium after UV irradiation. In mammals, Ro undergoes dramatic localization changes after UV irradiation, changing from mostly cytoplasmic to predominantly nuclear. Here, we report that a second role of Y RNAs is to regulate the subcellular distribution of Ro. A mutant Ro protein that does not bind Y RNAs accumulates in nuclei. Ro also localizes to nuclei when Y RNAs are depleted. By assaying chimeric proteins in which portions of mouse Ro were replaced with bacterial Ro sequences, we show that nuclear accumulation of Ro after irradiation requires sequences that overlap the Y RNA binding site. Ro also accumulates in nuclei after oxidative stress, and similar sequences are required. Together, these data reveal that Ro contains a signal for nuclear accumulation that is masked by a bound Y RNA and suggest that Y RNA binding may be modulated during cell stress.


Subject(s)
RNA, Untranslated/physiology , Ribonucleoproteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Nucleus/metabolism , Cells, Cultured , Deinococcus/genetics , Mice , Molecular Sequence Data , Oxidative Stress , Protein Structure, Tertiary , RNA Interference , RNA, Untranslated/metabolism , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Sequence Alignment
16.
Biochemistry ; 47(2): 762-70, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18092807

ABSTRACT

M1 RNA, the gene product of rnpB, is the catalytic subunit of RNase P in Escherichia coli. M1 RNA is transcribed from a proximal promoter as pM1 RNA, a precursor M1 RNA, and then is processed at its 3' end by RNase E. In addition to pM1 RNA, large rnpB-containing transcripts are produced from unknown upstream promoters. However, it is not known yet how these large transcripts contribute to M1 RNA biosynthesis. To examine their biological relevance to M1 RNA biosynthesis, we constructed a model upstream transcript, upRNA, and analyzed its cellular metabolism. We found that upRNA was primarily degraded rather than processed to M1 RNA in the cell and that this degradation occurred in RNase E-dependent manner. The in vitro cleavage assay with the N-terminal catalytic fraction of RNase E showed that the M1 RNA structural sequence in upRNA was much more vulnerable to the enzyme than the sequence in pM1 RNA. Considering that RNase E is a processing enzyme involved in 3' end formation of M1 RNA, our results imply that this enzyme plays a dual role in processing and degradation to achieve tight control of M1 RNA biosynthesis.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/biosynthesis , Escherichia coli/enzymology , Ribonuclease P/biosynthesis , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Gene Expression Regulation, Bacterial , Models, Biological , Nucleic Acid Conformation , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease P/chemistry , Substrate Specificity
17.
J Biol Chem ; 280(41): 34667-74, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16105832

ABSTRACT

M1 RNA, the catalytic subunit of Escherichia coli RNase P, is an essential ribozyme that processes the 5' leader sequence of precursor tRNAs. It is generated by the removal of 36 nucleotides from the 3' end of the primary rnpB transcript (pM1 RNA), but the biological significance of this reaction in bacterial metabolism remains obscure. In this study, we constructed and analyzed bacterial strains carrying mutations in the rne-dependent site of their rnpB genes, showing that the 3' processing of M1 RNA is essential for cell viability. Furthermore, we demonstrate that pM1 RNA can undergo not only 3' processing but also poly(A)-dependent degradation. Therefore, our results suggest that the 3' processing of M1 RNA provides a functional mechanism for the protection of its primary transcript against degradation.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Escherichia coli/enzymology , RNA, Messenger/chemistry , Ribonuclease P/genetics , Ribonuclease P/physiology , Base Sequence , Blotting, Northern , Catalysis , Cell Proliferation , Cell Survival , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Genotype , Models, Genetic , Molecular Sequence Data , Mutation , Nucleotides/chemistry , Oligonucleotides/chemistry , Phenotype , Plasmids/metabolism , Polynucleotide Adenylyltransferase/metabolism , RNA/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , Time Factors , Transgenes
18.
J Biochem ; 136(5): 693-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15632310

ABSTRACT

To gain insight into the mechanism by which the sequence at the rne-dependent site of substrate RNA affects the substrate specificity of Escherichia coli RNase E, we performed kinetic analysis of the cleavage of precursor M1 RNA molecules containing various sequences at the rne-dependent site by the N-terminal catalytic half of RNase E (NTH-RNase E). NTH-RNase E displayed higher K(m) and k(cat) values for more specific substrates. The retention of single strandedness at the rne-dependent site was essential for cleavage efficiency. Moreover, the loss of single-strandedness was accompanied by a decrease in both the K(m) and k(cat) values.


Subject(s)
Endoribonucleases/metabolism , RNA Precursors/metabolism , RNA, Catalytic/metabolism , Endoribonucleases/genetics , Escherichia coli/enzymology , Genetic Variation , Kinetics , Nucleic Acid Conformation , RNA Precursors/chemistry , RNA, Catalytic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Time Factors
19.
FEBS Lett ; 529(2-3): 225-31, 2002 Oct 09.
Article in English | MEDLINE | ID: mdl-12372605

ABSTRACT

M1 RNA, the catalytic component of Escherichia coli RNase P, is derived from the 3'-end processing of precursor M1 RNA, a major transcript of the rnpB gene. In this study, we investigated the mechanism of 3'-end processing of M1 RNA using the recombinant N-terminal half RNase E. The cleavage site preference of RNase E differed from that of the 40% ammonium sulfate precipitate (ASP-40), a partially purified cell extract containing processing activity. However, the addition of a trace amount of ASP-40 changed the cleavage site preference of RNase E to that of ASP-40 suggesting the involvement of a soluble factor in cleavage site preference.


Subject(s)
Endoribonucleases/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA/metabolism , Base Sequence , Chromatography, Gel , DNA Primers , Nucleic Acid Conformation , RNA/chemistry , RNA Precursors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...