Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Mol Cell ; 81(3): 514-529.e6, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33385327

ABSTRACT

Termination of RNA polymerase II (RNAPII) transcription in metazoans relies largely on the cleavage and polyadenylation (CPA) and integrator (INT) complexes originally found to act at the ends of protein-coding and small nuclear RNA (snRNA) genes, respectively. Here, we monitor CPA- and INT-dependent termination activities genome-wide, including at thousands of previously unannotated transcription units (TUs), producing unstable RNA. We verify the global activity of CPA occurring at pA sites indiscriminately of their positioning relative to the TU promoter. We also identify a global activity of INT, which is largely sequence-independent and restricted to a ~3-kb promoter-proximal region. Our analyses suggest two functions of genome-wide INT activity: it dampens transcriptional output from weak promoters, and it provides quality control of RNAPII complexes that are unfavorably configured for transcriptional elongation. We suggest that the function of INT in stable snRNA production is an exception from its general cellular role, the attenuation of non-productive transcription.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , DNA-Binding Proteins/metabolism , RNA Polymerase II/metabolism , RNA, Small Nuclear/biosynthesis , Transcription Termination, Genetic , Cleavage And Polyadenylation Specificity Factor/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Polyadenylation , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA, Small Nuclear/genetics
2.
Genes Dev ; 34(13-14): 989-1001, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32499401

ABSTRACT

Polymerases and exonucleases act on 3' ends of nascent RNAs to promote their maturation or degradation but how the balance between these activities is controlled to dictate the fates of cellular RNAs remains poorly understood. Here, we identify a central role for the human DEDD deadenylase TOE1 in distinguishing the fates of small nuclear (sn)RNAs of the spliceosome from unstable genome-encoded snRNA variants. We found that TOE1 promotes maturation of all regular RNA polymerase II transcribed snRNAs of the major and minor spliceosomes by removing posttranscriptional oligo(A) tails, trimming 3' ends, and preventing nuclear exosome targeting. In contrast, TOE1 promotes little to no maturation of tested U1 variant snRNAs, which are instead targeted by the nuclear exosome. These observations suggest that TOE1 is positioned at the center of a 3' end quality control pathway that selectively promotes maturation and stability of regular snRNAs while leaving snRNA variants unprocessed and exposed to degradation in what could be a widespread mechanism of RNA quality control given the large number of noncoding RNAs processed by DEDD deadenylases.


Subject(s)
Nuclear Proteins/metabolism , RNA 3' End Processing/genetics , RNA Stability/genetics , RNA, Small Nuclear/genetics , Cell Line , Cell Nucleus/metabolism , Gene Deletion , HeLa Cells , Humans , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , RNA, Small Nuclear/biosynthesis
3.
Genes (Basel) ; 10(11)2019 11 18.
Article in English | MEDLINE | ID: mdl-31752243

ABSTRACT

In the ciliate Stylonychia, somatic macronuclei differentiate from germline micronuclei during sexual reproduction, accompanied by developmental sequence reduction. Concomitantly, over 95% of micronuclear sequences adopt a heterochromatin structure characterized by the histone variant H3.4 and H3K27me3. RNAi-related genes and histone variants dominate the list of developmentally expressed genes. Simultaneously, 27nt-ncRNAs that match sequences retained in new macronuclei are synthesized and bound by PIWI1. Recently, we proposed a mechanistic model for 'RNA-induced DNA replication interference' (RIRI): during polytene chromosome formation PIWI1/27nt-RNA-complexes target macronucleus-destined sequences (MDS) by base-pairing and temporarily cause locally stalled replication. At polytene chromosomal segments with ongoing replication, H3.4K27me3-nucleosomes become selectively deposited, thus dictating the prospective heterochromatin structure of these areas. Consequently, these micronucleus-specific sequences become degraded, whereas 27nt-RNA-covered sites remain protected. However, the biogenesis of the 27nt-RNAs remains unclear. It was proposed earlier that in stichotrichous ciliates 27nt-RNA precursors could derive from telomere-primed bidirectional transcription of nanochromosomes and subsequent Dicer-like (DCL) activity. As a minimalistic explanation, we propose here that the 27nt-RNA precursor could rather be mRNA or pre-mRNA and that the transition of coding RNA from parental macronuclei to non-coding RNAs, which act in premature developing macronuclei, could involve RNA-dependent RNA polymerase (RDRP) activity creating dsRNA intermediates prior to a DCL-dependent pathway. Interestingly, by such mechanism the partition of a parental somatic genome and possibly also the specific nanochromosome copy numbers could be vertically transmitted to the differentiating nuclei of the offspring.


Subject(s)
Ciliophora/genetics , Gene Expression Regulation, Developmental , Micronucleus, Germline/genetics , RNA, Messenger/biosynthesis , RNA, Small Nuclear/biosynthesis , DNA Replication , Genome, Protozoan/genetics , Histones/genetics , Histones/metabolism , Micronucleus, Germline/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , RNA Interference , RNA Precursors/biosynthesis , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Small Nuclear/genetics , Telomere/genetics , Telomere/metabolism
4.
Biomed Res Int ; 2019: 1425281, 2019.
Article in English | MEDLINE | ID: mdl-31058184

ABSTRACT

Leishmania major, a protozoan parasite that diverged early from the main eukaryotic lineage, exhibits unusual mechanisms of gene expression. Little is known in this organism about the transcription factors involved in the synthesis of tRNA, 5S rRNA, and snRNAs, transcribed by RNA Polymerase III (Pol III). Here we identify and characterize the TFIIIB subunit Bdp1 in L. major (LmBdp1). Bdp1 plays key roles in Pol III transcription initiation in other organisms, as it participates in Pol III recruitment and promoter opening. In silico analysis showed that LmBdp1 contains the typical extended SANT domain as well as other Bdp1 conserved regions. Nevertheless, LmBdp1 also displays distinctive features, including the presence of only one aromatic residue in the N-linker region. We were not able to produce null mutants of LmBdp1 by homologous recombination, as the obtained double replacement cell line contained an extra copy of LmBdp1, indicating that LmBdp1 is essential for the viability of L. major promastigotes. Notably, the mutant cell line showed reduced levels of the LmBdp1 protein, and its growth was significantly decreased in relation to wild-type cells. Nuclear run-on assays demonstrated that Pol III transcription was affected in the mutant cell line, and ChIP experiments showed that LmBdp1 binds to 5S rRNA, tRNA, and snRNA genes. Thus, our results indicate that LmBdp1 is an essential protein required for Pol III transcription in L. major.


Subject(s)
Leishmania major/genetics , RNA Polymerase III/genetics , Transcription Factor TFIIIB/genetics , Transcription, Genetic , Computer Simulation , Conserved Sequence/genetics , Gene Expression Regulation/genetics , Homologous Recombination/genetics , Mutant Proteins/genetics , Promoter Regions, Genetic , Protein Domains/genetics , Protein Subunits/genetics , RNA, Ribosomal, 5S/biosynthesis , RNA, Small Nuclear/biosynthesis , RNA, Transfer/biosynthesis
5.
Nat Chem Biol ; 15(4): 340-347, 2019 04.
Article in English | MEDLINE | ID: mdl-30778204

ABSTRACT

Small nuclear RNAs (snRNAs) are core spliceosome components and mediate pre-mRNA splicing. Here we show that snRNAs contain a regulated and reversible nucleotide modification causing them to exist as two different methyl isoforms, m1 and m2, reflecting the methylation state of the adenosine adjacent to the snRNA cap. We find that snRNA biogenesis involves the formation of an initial m1 isoform with a single-methylated adenosine (2'-O-methyladenosine, Am), which is then converted to a dimethylated m2 isoform (N6,2'-O-dimethyladenosine, m6Am). The relative m1 and m2 isoform levels are determined by the RNA demethylase FTO, which selectively demethylates the m2 isoform. We show FTO is inhibited by the oncometabolite D-2-hydroxyglutarate, resulting in increased m2-snRNA levels. Furthermore, cells that exhibit high m2-snRNA levels show altered patterns of alternative splicing. Together, these data reveal that FTO controls a previously unknown central step of snRNA processing involving reversible methylation, and suggest that epitranscriptomic information in snRNA may influence mRNA splicing.


Subject(s)
Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , RNA, Small Nuclear/biosynthesis , Adenosine/biosynthesis , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alternative Splicing , Animals , HEK293 Cells , Humans , Male , Methylation , Mice , Mice, Knockout , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Nuclear/metabolism
6.
RNA ; 24(4): 437-460, 2018 04.
Article in English | MEDLINE | ID: mdl-29367453

ABSTRACT

Removal of introns from precursor messenger RNA (pre-mRNA) and some noncoding transcripts is an essential step in eukaryotic gene expression. In the nucleus, this process of RNA splicing is carried out by the spliceosome, a multi-megaDalton macromolecular machine whose core components are conserved from yeast to humans. In addition to many proteins, the spliceosome contains five uridine-rich small nuclear RNAs (snRNAs) that undergo an elaborate series of conformational changes to correctly recognize the splice sites and catalyze intron removal. Decades of biochemical and genetic data, along with recent cryo-EM structures, unequivocally demonstrate that U6 snRNA forms much of the catalytic core of the spliceosome and is highly dynamic, interacting with three snRNAs, the pre-mRNA substrate, and >25 protein partners throughout the splicing cycle. This review summarizes the current state of knowledge on how U6 snRNA is synthesized, modified, incorporated into snRNPs and spliceosomes, recycled, and degraded.


Subject(s)
RNA Splice Sites/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Small Nuclear/genetics , Spliceosomes/metabolism , Humans , Nucleic Acid Conformation , RNA Polymerase III/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA, Small Nuclear/biosynthesis , Saccharomyces cerevisiae/genetics
7.
Plant Cell ; 29(12): 3214-3233, 2017 12.
Article in English | MEDLINE | ID: mdl-29093215

ABSTRACT

Phosphorylation of the RNA polymerase II (Pol II) C-terminal domain (CTD) regulates transcription of protein-coding mRNAs and noncoding RNAs. CTD function in transcription of protein-coding RNAs has been studied extensively, but its role in plant noncoding RNA transcription remains obscure. Here, using Arabidopsis thaliana CTD PHOSPHATASE-LIKE4 knockdown lines (CPL4RNAi ), we showed that CPL4 functions in genome-wide, conditional production of 3'-extensions of small nuclear RNAs (snRNAs) and biogenesis of novel transcripts from protein-coding genes downstream of the snRNAs (snRNA-downstream protein-coding genes [snR-DPGs]). Production of snR-DPGs required the Pol II snRNA promoter (PIIsnR), and CPL4RNAi plants showed increased read-through of the snRNA 3'-end processing signal, leading to continuation of transcription downstream of the snRNA gene. We also discovered an unstable, intermediate-length RNA from the SMALL SCP1-LIKE PHOSPHATASE14 locus (imRNASSP14 ), whose expression originated from the 5' region of a protein-coding gene. Expression of the imRNASSP14 was driven by a PIIsnR and was conditionally 3'-extended to produce an mRNA. In the wild type, salt stress induced the snRNA-to-snR-DPG switch, which was associated with alterations of Pol II-CTD phosphorylation at the target loci. The snR-DPG transcripts occur widely in plants, suggesting that the transcriptional snRNA-to-snR-DPG switch may be a ubiquitous mechanism to regulate plant gene expression in response to environmental stresses.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/physiology , Phosphoprotein Phosphatases/metabolism , RNA, Messenger/biosynthesis , RNA, Small Nuclear/biosynthesis , Salt Stress/physiology , Arabidopsis/genetics , DNA Transposable Elements/genetics , Gene Expression Regulation, Plant/drug effects , Genes, Plant , Genetic Loci , Luciferases/metabolism , Models, Biological , Mutation/genetics , Nucleotide Motifs/genetics , Open Reading Frames/genetics , Phosphorylation , Plants, Genetically Modified , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Plant/metabolism , RNA, Small Nuclear/genetics , Transcription Factors/metabolism , Up-Regulation/genetics
8.
Parasit Vectors ; 10(1): 281, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-28583151

ABSTRACT

BACKGROUND: RNA polymerase III promoters have been widely used to express short hairpin-RNA (shRNA), microRNA (miRNA), and small guide RNA (sgRNA) in gene functional analysis in a variety of organisms including Schistosoma mansoni. However, no endogenous RNA polymerase III promoters have been identified in Schistosoma japonicum. The lack of appropriate promoters in S. japonicum has hindered its gene functional analysis. Identification of functional promoters in S. japonicum is therefore in urgent need. RESULTS: Via sequence alignment, a 347 bp sequence upstream from the coding region of S. japonicum U6 small nuclear RNA (snRNA) was identified, cloned, and named as S. japonicum U6 (sjU6) promoter. A sgRNA sequence named as sgRNA970 was designed, and its Cas9 nuclease guiding activity was confirmed by in vitro cleavage assay. The sjU6 promoter was ligated with sgRNA970 coding sequence by overlap PCR to generate a sjU6-sgRNA970 expression cassette. The expression cassette was inserted into a lentiviral plasmid to construct the pHBLV-sgRNA970 plasmid. First, we tested the sjU6 promoter activity in HEK293 cells by transfecting HEK293 cells with the pHBLV-sgRNA970 plasmid. RT-PCR amplification of the total RNA from the transfected HEK293 cells confirmed the presence of sgRNA970 transcript and indicated sjU6 promoter was functional to initiate transcription in HEK293 cells. Then we transduced the lentivirus expressing Cas9-ZsGreen fusion protein into 14 dpi schistosomula to test whether lentivirus was capable to induce exogenous gene expression in S. japonicum. Fluorescence microscopy and western blot results confirmed the expression of Cas9-ZsGreen fusion protein in S. japonicum. Therefore, this lentiviral system was adapted to test promoter activity in S. japonicum. Finally, we transduced 14 dpi S. japonicum with lentivirus produced from the pHBLV-sgRNA970 plasmid. RT-PCR amplification of the total RNA from transduced schistosomula confirmed the presence of sgRNA970 transcript and therefore indicated sjU6 promoter was functional to initiate transcription in S. japonicum. CONCLUSION: To our knowledge, sjU6 promoter would be the first identified and validated endogenous RNA polymerase III promoter in S. japonicum, which could be used for future CRISPR/Cas9 studies in S. japonicum.


Subject(s)
Promoter Regions, Genetic , RNA Polymerase III/metabolism , RNA, Small Nuclear/biosynthesis , RNA, Small Nuclear/genetics , Schistosoma japonicum/genetics , Animals , Computational Biology , Gene Expression , Gene Expression Profiling , HEK293 Cells , Humans , Lentivirus/genetics , Plasmids , Recombination, Genetic
9.
J R Soc Interface ; 14(130)2017 05.
Article in English | MEDLINE | ID: mdl-28468917

ABSTRACT

The circadian regulation of gene expression allows plants and animals to anticipate predictable environmental changes. While the influence of the circadian clock has recently been shown to extend to ribosome biogenesis, the dynamics and regulation of the many small nucleolar RNA that are required in pre-ribosomal RNA folding and modification are unknown. Using a novel computational method, we show that 18S and 28S pre-rRNA are subject to circadian regulation in a nuclear RNA sequencing time course. A population of snoRNA with circadian expression is identified that is functionally associated with rRNA modification. More generally, we find the abundance of snoRNA known to modify 18S and 28S to be inversely correlated with the abundance of their target. Cyclic patterns in the expression of a number of snoRNA indicate a coordination with rRNA maturation, potentially through an upregulation in their biogenesis, or their release from mature rRNA at the end of the previous cycle of rRNA maturation, in antiphase with the diurnal peak in pre-rRNA. Few cyclic snoRNA have cyclic host genes, indicating the action of regulatory mechanisms in addition to transcriptional activation of the host gene. For highly expressed independently transcribed snoRNA, we find a characteristic RNA polymerase II and H3K4me3 signature that correlates with mean snoRNA expression over the day.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Liver/metabolism , Models, Biological , RNA, Small Nuclear/biosynthesis , Animals , Mice , RNA, Ribosomal, 18S/biosynthesis , RNA, Ribosomal, 28S/biosynthesis
10.
EMBO J ; 36(7): 934-948, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28254838

ABSTRACT

The 7SK small nuclear RNP (snRNP), composed of the 7SK small nuclear RNA (snRNA), MePCE, and Larp7, regulates the mRNA elongation capacity of RNA polymerase II (RNAPII) through controlling the nuclear activity of positive transcription elongation factor b (P-TEFb). Here, we demonstrate that the human 7SK snRNP also functions as a canonical transcription factor that, in collaboration with the little elongation complex (LEC) comprising ELL, Ice1, Ice2, and ZC3H8, promotes transcription of RNAPII-specific spliceosomal snRNA and small nucleolar RNA (snoRNA) genes. The 7SK snRNA specifically associates with a fraction of RNAPII hyperphosphorylated at Ser5 and Ser7, which is a hallmark of RNAPII engaged in snRNA synthesis. Chromatin immunoprecipitation (ChIP) and chromatin isolation by RNA purification (ChIRP) experiments revealed enrichments for all components of the 7SK snRNP on RNAPII-specific sn/snoRNA genes. Depletion of 7SK snRNA or Larp7 disrupts LEC integrity, inhibits RNAPII recruitment to RNAPII-specific sn/snoRNA genes, and reduces nascent snRNA and snoRNA synthesis. Thus, through controlling both mRNA elongation and sn/snoRNA synthesis, the 7SK snRNP is a key regulator of nuclear RNA production by RNAPII.


Subject(s)
Gene Expression Regulation , RNA, Small Nuclear/biosynthesis , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , Chromatin Immunoprecipitation , HeLa Cells , Humans , RNA Polymerase II/metabolism
11.
Parasit Vectors ; 9(1): 401, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27430335

ABSTRACT

BACKGROUND: Leishmania and other trypanosomatid parasites possess atypical mechanisms of gene expression, including the maturation of mRNAs by trans-splicing and the involvement of RNA Polymerase III in transcription of all snRNA molecules. Since snRNAs are essential for trans-splicing, we are interested in the study of the sequences that direct their expression. Here we report the characterization of L. major U2 snRNA promoter region. RESULTS: All species of Leishmania possess a single U2 snRNA gene that contains a divergently-oriented tRNA-Ala gene in the upstream region. Between these two genes we found a tRNA-like sequence that possesses conserved boxes A and B. Primer extension and RT-qPCR analyses with RNA from transiently-transfected cells showed that transcription of L. major U2 snRNA is almost abolished when boxes A and B from the tRNA-like are deleted or mutated. The levels of the U2 snRNA were also highly affected when base substitutions were introduced into box B from the tRNA-Ala gene and the first nucleotides of the U2 snRNA gene itself. We also demonstrate that the tRNA-like is transcribed, generating a main transcript of around 109 bases. As pseudouridines in snRNAs are required for splicing in other organisms, we searched for this modified nucleotide in the L. major U2 snRNA. Our results show the presence of six pseudouridines in the U2 snRNA, including one in the Sm site that has not been reported in other organisms. CONCLUSIONS: Four different regions control the transcription of the U2 snRNA gene in L. major: boxes A and B from the neighbor tRNA-like, box B from the upstream tRNA-Ala gene and the first nucleotides of the U2 snRNA. Thus, the promoter region of L. major U2 snRNA is different from any other promoter reported for snRNAs. Pseudouridines could play important roles in L. major U2 snRNA, since they were found in functionally important regions, including the branch point recognition region and the Sm binding site.


Subject(s)
Leishmania major/genetics , Promoter Regions, Genetic , RNA, Small Nuclear/biosynthesis , RNA, Transfer, Ala/genetics , Transcription, Genetic , DNA Mutational Analysis , Pseudouridine/analysis , RNA, Small Nuclear/chemistry
12.
PLoS One ; 10(9): e0137398, 2015.
Article in English | MEDLINE | ID: mdl-26340001

ABSTRACT

Colletotrichum lentis is a fungal pathogen of lentil in Canada but rarely reported elsewhere. Two races, Ct0 and Ct1, have been identified using differential lines. Our objective was to develop a PCR-probe differentiating these races. Sequences of the translation elongation factor 1α (tef1α), RNA polymerase II subunit B2 (rpb2), ATP citrate lyase subunit A (acla), and internal transcribed spacer (ITS) regions were monomorphic, while the intergenic spacer (IGS) region showed length polymorphisms at two minisatellites of 23 and 39 nucleotides (nt). A PCR-probe (39F/R) amplifying the 39 nt minisatellite was developed which subsequently revealed 1-5 minisatellites with 1-12 repeats in C. lentis. The probe differentiated race Ct1 isolates having 7, 9 or 7+9 repeats from race Ct0 having primarily 2 or 4 repeats, occasionally 5, 6, or 8, but never 7 or 9 repeats. These isolates were collected between 1991 and 1999. In a 2012 survey isolates with 2 and 4 repeats increased from 34% to 67%, while isolated with 7 or 9 repeats decreased from 40 to 4%, likely because Ct1 resistant lentil varieties had been grown. The 39 nt repeat was identified in C. gloeosporioides, C. trifolii, Ascochyta lentis, Sclerotinia sclerotiorum and Botrytis cinerea. Thus, the 39F/R PCR probe is not species specific, but can differentiate isolates based on repeat number. The 23 nt minisatellite in C. lentis exists as three length variants with ten sequence variations differentiating race Ct0 having 14 or 19 repeats from race Ct1 having 17 repeats, except for one isolate. RNA-translation of 23 nt repeats forms hairpins and has the appropriate length to suggest that IGS could be a site of small RNA synthesis, a hypothesis that warrants further investigation. Small RNA from fungal plant pathogens able to silence genes either in the host or pathogen thereby aiding infection have been reported.


Subject(s)
Colletotrichum/pathogenicity , Eukaryotic Initiation Factors/genetics , Fungal Proteins/genetics , Minisatellite Repeats , Polymorphism, Genetic , RNA, Small Nuclear/genetics , ATP Citrate (pro-S)-Lyase/genetics , Ascomycota/genetics , Ascomycota/pathogenicity , Base Sequence , Botrytis/genetics , Botrytis/pathogenicity , Colletotrichum/genetics , DNA Probes , DNA, Ribosomal Spacer/genetics , Host-Pathogen Interactions , Lens Plant/microbiology , Molecular Sequence Data , Mycological Typing Techniques , Plant Diseases/microbiology , Protein Subunits/genetics , RNA Polymerase II/genetics , RNA, Small Nuclear/biosynthesis , Sequence Analysis, DNA , Species Specificity , Virulence
13.
PLoS One ; 10(4): e0122348, 2015.
Article in English | MEDLINE | ID: mdl-25875178

ABSTRACT

Small nuclear ribonucleoproteins (snRNPs), which are required for pre-mRNA splicing, contain extensively modified snRNA. Small Cajal body-specific ribonucleoproteins (scaRNPs) mediate these modifications. It is unknown how the box C/D class of scaRNPs localizes to Cajal Bodies (CBs). The processing of box C/D scaRNA is also unclear. Here, we explore the processing of box C/D scaRNA 2 and 9 by coilin. We also broaden our investigation to include WRAP53 and SMN, which accumulate in CBs, play a role in RNP biogenesis and associate with coilin. These studies demonstrate that the processing of an ectopically expressed scaRNA2 is altered upon the reduction of coilin, WRAP53 or SMN, but the extent and direction of this change varies depending on the protein reduced. We also show that box C/D scaRNP activity is reduced in a cell line derived from coilin knockout mice. Collectively, the findings presented here further implicate coilin as being a direct participant in the formation of box C/D scaRNPs, and demonstrate that WRAP53 and SMN may also play a role, but the activity of these proteins is divergent to coilin.


Subject(s)
Coiled Bodies/metabolism , RNA Splicing/genetics , RNA, Small Nuclear/biosynthesis , Ribonucleoproteins, Small Nuclear/biosynthesis , Animals , Coiled Bodies/genetics , Cyclic AMP Response Element-Binding Protein/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Molecular Chaperones , RNA Precursors/genetics , RNA, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/genetics , Telomerase/genetics
14.
PLoS One ; 9(5): e93846, 2014.
Article in English | MEDLINE | ID: mdl-24796678

ABSTRACT

BACKGROUND: Gene expression studies of peripheral blood mononuclear cells from patients with systemic lupus erythematosus (SLE) have demonstrated a type I interferon signature and increased expression of inflammatory cytokine genes. Studies of patients with Aicardi Goutières syndrome, commonly cited as a single gene model for SLE, have suggested that accumulation of non-coding RNAs may drive some of the pathologic gene expression, however, no RNA sequencing studies of SLE patients have been performed. This study was designed to define altered expression of coding and non-coding RNAs and to detect globally altered RNA processing in SLE. METHODS: Purified monocytes from eight healthy age/gender matched controls and nine SLE patients (with low-moderate disease activity and lack of biologic drug use or immune suppressive treatment) were studied using RNA-seq. Quantitative RT-PCR was used to validate findings. Serum levels of endotoxin were measured by ELISA. RESULTS: We found that SLE patients had diminished expression of most endogenous retroviruses and small nucleolar RNAs, but exhibited increased expression of pri-miRNAs. Splicing patterns and polyadenylation were significantly altered. In addition, SLE monocytes expressed novel transcripts, an effect that was replicated by LPS treatment of control monocytes. We further identified increased circulating endotoxin in SLE patients. CONCLUSIONS: Monocytes from SLE patients exhibit globally dysregulated gene expression. The transcriptome is not simply altered by the transcriptional activation of a set of genes, but is qualitatively different in SLE. The identification of novel loci, inducible by LPS, suggests that chronic microbial translocation could contribute to the immunologic dysregulation in SLE, a new potential disease mechanism.


Subject(s)
Endotoxins/blood , Gene Expression Regulation , Lupus Erythematosus, Systemic/metabolism , Monocytes/metabolism , RNA, Small Nuclear/biosynthesis , Transcriptome , Female , Humans , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/pathology , Male , Monocytes/pathology , RNA Precursors
15.
Mol Cell Biol ; 34(13): 2488-98, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752900

ABSTRACT

The C-terminal domain of the RNA polymerase II largest subunit (the Rpb1 CTD) is composed of tandem heptad repeats of the consensus sequence Y(1)S(2)P(3)T(4)S(5)P(6)S(7). We reported previously that Thr 4 is phosphorylated and functions in histone mRNA 3'-end formation in chicken DT40 cells. Here, we have extended our studies on Thr 4 and to other CTD mutations by using these cells. We found that an Rpb1 derivative containing only the N-terminal half of the CTD, as well as a similar derivative containing all-consensus repeats (26r), conferred full viability, while the C-terminal half, with more-divergent repeats, did not, reflecting a strong and specific defect in snRNA 3'-end formation. Mutation in 26r of all Ser 2 (S2A) or Ser 5 (S5A) residues resulted in lethality, while Ser 7 (S7A) mutants were fully viable. While S2A and S5A cells displayed defects in transcription and RNA processing, S7A cells behaved identically to 26r cells in all respects. Finally, we found that Thr 4 was phosphorylated by cyclin-dependent kinase 9 in cells and dephosphorylated both in vitro and in vivo by the phosphatase Fcp1.


Subject(s)
Cyclin-Dependent Kinase 9/genetics , Phosphoprotein Phosphatases/genetics , RNA Polymerase II/genetics , RNA Processing, Post-Transcriptional , Threonine/chemistry , Amino Acid Sequence , Animals , Cell Line , Chickens , HEK293 Cells , Humans , Mutation , Phosphorylation/genetics , Protein Subunits/genetics , RNA, Small Nuclear/biosynthesis , RNA, Small Nuclear/genetics , Threonine/genetics , Transcription, Genetic
16.
Nucleic Acids Res ; 42(1): 264-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097444

ABSTRACT

RNA polymerase II transcribes both protein coding and non-coding RNA genes and, in yeast, different mechanisms terminate transcription of the two gene types. Transcription termination of mRNA genes is intricately coupled to cleavage and polyadenylation, whereas transcription of small nucleolar (sno)/small nuclear (sn)RNA genes is terminated by the RNA-binding proteins Nrd1, Nab3 and Sen1. The existence of an Nrd1-like pathway in humans has not yet been demonstrated. Using the U1 and U2 genes as models, we show that human snRNA genes are more similar to mRNA genes than yeast snRNA genes with respect to termination. The Integrator complex substitutes for the mRNA cleavage and polyadenylation specificity factor complex to promote cleavage and couple snRNA 3'-end processing with termination. Moreover, members of the associated with Pta1 (APT) and cleavage factor I/II complexes function as transcription terminators for human snRNA genes with little, if any, role in snRNA 3'-end processing. The gene-specific factor, proximal sequence element-binding transcription factor (PTF), helps clear the U1 and U2 genes of nucleosomes, which provides an easy passage for pol II, and the negative elongation factor facilitates termination at the end of the genes where nucleosome levels increase. Thus, human snRNA genes may use chromatin structure as an additional mechanism to promote efficient transcription termination in vivo.


Subject(s)
RNA, Small Nuclear/genetics , Transcription Termination, Genetic , mRNA Cleavage and Polyadenylation Factors/metabolism , Chromatin/chemistry , HeLa Cells , Humans , RNA 3' End Processing , RNA, Small Nuclear/biosynthesis , RNA, Small Nuclear/metabolism , Transcription Factors/physiology
17.
J Biol Chem ; 288(38): 27564-27570, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23955442

ABSTRACT

In the fruit fly Drosophila melanogaster, RNA polymerase III transcription was found to be dependent not upon the canonical TATA box-binding protein (TBP) but instead upon the TBP-related factor 1 (TRF1) (Takada, S., Lis, J. T., Zhou, S., and Tjian, R. (2000) Cell 101, 459-469). Here we confirm that transcription of fly tRNA genes requires TRF1. However, we unexpectedly find that U6 snRNA gene promoters are occupied primarily by TBP in cells and that knockdown of TBP, but not TRF1, inhibits U6 transcription in cells. Moreover, U6 transcription in vitro effectively utilizes TBP, whereas TBP cannot substitute for TRF1 to promote tRNA transcription in vitro. Thus, in fruit flies, different classes of RNA polymerase III promoters differentially utilize TBP and TRF1 for the initiation of transcription.


Subject(s)
Drosophila Proteins/metabolism , Promoter Regions, Genetic/physiology , RNA Polymerase III/metabolism , TATA Box Binding Protein-Like Proteins/metabolism , TATA-Box Binding Protein/metabolism , Transcription, Genetic/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , RNA Polymerase III/genetics , RNA, Small Nuclear/biosynthesis , RNA, Small Nuclear/genetics , RNA, Transfer/biosynthesis , RNA, Transfer/genetics , TATA Box Binding Protein-Like Proteins/genetics , TATA-Box Binding Protein/genetics
18.
Wiley Interdiscip Rev RNA ; 4(5): 581-92, 2013.
Article in English | MEDLINE | ID: mdl-23776162

ABSTRACT

U6 snRNA is one of five uridine-rich noncoding RNAs that form the major spliceosome complex. Unlike other U-snRNAs, it reveals many distinctive aspects of biogenesis such as transcription by RNA polymerase III, transcript nuclear retention and particular features of transcript ends: monomethylated 5'-guanosine triphosphate as cap structure and a 2',3'-cyclic phosphate moiety (>P) at the 3' termini. U6-snRNA plays a central role in splicing and thus its transcription, maturation, snRNP formation, and recycling are essential for cellular homeostasis. U6 snRNA enters the splicing cycle as part of the tri-U4/U6.U5snRNP complex, and after significant structural arrangements forms the catalytic site of the spliceosome together with U2 snRNA and Prp8. U6 snRNA also contributes to the splicing reaction by coordinating metal cations required for catalysis. Many human diseases are associated with altered splicing processes. Disruptions of the basal splicing machinery can be lethal or lead to severe diseases such as spinal muscular atrophy, amyotrophic lateral sclerosis, or retinitis pigmentosa. Recent studies have identified a new U6 snRNA biogenesis factor Usb1, the absence of which leads to poikiloderma with neutropenia (PN) (OMIM 604173), an autosomal recessive skin disease. Usb1 is an evolutionarily conserved 3'→5' exoribonuclease that is responsible for removing 3'-terminal uridines from U6 snRNA transcripts, which leads to the formation of a 2',3' cyclic phosphate moiety (>P). This maturation step is fundamental for U6 snRNP assembly and recycling. Usb1 represents the first example of a direct association between a spliceosomal U6 snRNA biogenesis factor and human genetic disease.


Subject(s)
Genetic Diseases, Inborn/genetics , RNA Splicing , RNA, Small Nuclear/biosynthesis , Transcription, Genetic , Amyotrophic Lateral Sclerosis/genetics , Humans , Muscular Atrophy, Spinal/genetics , Neutropenia/genetics , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/metabolism , RNA, Small Nuclear/chemistry , Retinitis Pigmentosa/genetics , Skin Abnormalities/genetics
19.
Wiley Interdiscip Rev RNA ; 4(5): 491-506, 2013.
Article in English | MEDLINE | ID: mdl-23754627

ABSTRACT

The release of nascent RNA from transcribing RNA polymerase complexes is required for all further functions carried out by RNA molecules. The elements and processing machinery involved in 3' end formation therefore represent key determinants in the biogenesis and accumulation of cellular RNA. While these factors have been well-characterized for messenger RNA, recent work has elucidated analogous pathways for the 3' end formation of other important cellular RNA. Here, we discuss four specific cases of non-mRNA 3' end formation-metazoan small nuclear RNA, Saccharomyces cerevisiae small nuclear RNA, Schizosaccharomyces pombe telomerase RNA, and the mammalian MALAT1 large noncoding RNA-as models of alternative mechanisms to generate RNA 3' ends. Comparison of these disparate processing pathways reveals an emerging theme of evolutionary ingenuity. In some instances, evidence for the creation of a dedicated processing complex exists; while in others, components are utilized from the existing RNA processing machinery and modified to custom fit the unique needs of the RNA substrate. Regardless of the details of how non-mRNA 3' ends are formed, the lengths to which biological systems will go to release nascent transcripts from their DNA templates are fundamental for cell survival.


Subject(s)
RNA, Long Noncoding/biosynthesis , RNA, Small Nuclear/biosynthesis , RNA/biosynthesis , Telomerase/biosynthesis , Humans , Metabolic Networks and Pathways , Models, Biological , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism
20.
Plant Signal Behav ; 8(8)2013 Aug.
Article in English | MEDLINE | ID: mdl-23673353

ABSTRACT

We recently reported that hairpin (or stem-loop) priming is better-suited than polyA tailing to generate cDNA for plant microRNA qPCR. One major limitation of this method is the need to perform individual cDNA synthesis reactions for the reference gene and test miRNAs. Here, we report a novel fusion primer that allows multiplexed hairpin cDNA synthesis of the most-commonly used reference gene, nucleolar small RNA U6, together with test miRNAs. We also propose the use of miR1515 as a house keeping control for tropical legumes. We show that multiplexed cDNA synthesis does not result in loss of sensitivity and reduces the amount of RNA required for miRNA gene expression assays.


Subject(s)
DNA, Complementary/chemistry , DNA, Complementary/metabolism , MicroRNAs/biosynthesis , Nucleic Acid Conformation , RNA, Small Nuclear/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Gene Expression Regulation, Plant , Plants/genetics , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...