Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J BUON ; 26(5): 2084-2089, 2021.
Article in English | MEDLINE | ID: mdl-34761620

ABSTRACT

PURPOSE: To analyze the influence of DDX46 on the proliferative and migratory potentials of glioblastoma (GBM). METHODS: Differential levels of DDX46 in GBM cases and controls were examined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. By intervening DDX46 in U87 and U251 cells, proliferative and migratory changes were determined by colony formation assay, 5-Ethynyl-2'- deoxyuridine (EdU) assay and Transwell assay, respectively. Protein levels of p-p38, p38, cyclin D1 and MMP7 in GBM cells intervened by DDX46 or the inhibitor of p38 MAPK were detected. RESULTS: DDX46 was upregulated in GBM cases. Knockdown of DDX46 attenuated the proliferative capacity of GBM cells, and its overexpression enhanced the proliferative rate. The migratory capacity of GBM was not affected by DDX46. Overexpression of DDX46 upregulated p-p38 and cyclin D1 in GBM cells. The regulatory effect of DDX46 on GBM proliferation could be partially reversed by the treatment of doramapimod. CONCLUSIONS: DDX46 is upregulated in GBM, which strengthens the proliferative capacity of GBM by activating the MAPK-p38 signaling.


Subject(s)
Cell Movement , Cell Proliferation , DEAD-box RNA Helicases/physiology , Glioblastoma/pathology , MAP Kinase Signaling System/physiology , Ribonucleoprotein, U2 Small Nuclear/physiology , Humans , Time Factors , Tumor Cells, Cultured
2.
J Exp Bot ; 71(3): 751-758, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31605606

ABSTRACT

Flowering transition is regulated by complex genetic networks in response to endogenous and environmental signals. Pre-mRNA splicing is an essential step for the post-transcriptional regulation of gene expression. Alternative splicing of key flowering genes has been investigated in detail over the past decade. However, few splicing factors have been identified as being involved in flowering transition. Human heterodimeric splicing factor U2 snRNP auxiliary factor (U2AF) consists of two subunits, U2AF35 and U2AF65, and functions in 3' splice site recognition in mRNA splicing. Recent studies reveal that Arabidopsis U2AF65a/b and U2AF35a/b play important roles in the splicing of key flowering genes. We summarize recent advances in research on splicing-regulated flowering transition by focusing on the role of Arabidopsis U2AF in the splicing of key flowering-related genes at ambient temperature and in the abscisic acid signaling pathways.


Subject(s)
Alternative Splicing , Arabidopsis Proteins/metabolism , Flowers/physiology , MADS Domain Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/physiology , Abscisic Acid/metabolism , Arabidopsis
3.
Br J Haematol ; 171(4): 478-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26255870

ABSTRACT

Refractory anaemia with ring sideroblasts (RARS) is distinguished by hyperplastic inefficient erythropoiesis, aberrant mitochondrial ferritin accumulation and anaemia. Heterozygous mutations in the spliceosome gene SF3B1 are found in a majority of RARS cases. To explore the link between SF3B1 mutations and anaemia, we studied mutated RARS CD34(+) marrow cells with regard to transcriptome sequencing, splice patterns and mutational allele burden during erythroid differentiation. Transcriptome profiling during early erythroid differentiation revealed a marked up-regulation of genes involved in haemoglobin synthesis and in the oxidative phosphorylation process, and down-regulation of mitochondrial ABC transporters compared to normal bone marrow. Moreover, mis-splicing of genes involved in transcription regulation, particularly haemoglobin synthesis, was confirmed, indicating a compromised haemoglobinization during RARS erythropoiesis. In order to define the phase during which erythroid maturation of SF3B1 mutated cells is most affected, we assessed allele burden during erythroid differentiation in vitro and in vivo and found that SF3B1 mutated erythroblasts showed stable expansion until late erythroblast stage but that terminal maturation to reticulocytes was significantly reduced. In conclusion, SF3B1 mutated RARS progenitors display impaired splicing with potential downstream consequences for genes of key importance for haemoglobin synthesis and terminal erythroid differentiation.


Subject(s)
Anemia, Refractory/genetics , Anemia, Sideroblastic/genetics , Erythropoiesis/genetics , Hemoglobins/biosynthesis , Phosphoproteins/genetics , RNA Splicing/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Aged , Aged, 80 and over , Anemia, Refractory/blood , Anemia, Sideroblastic/blood , Biological Transport/genetics , Gene Expression Profiling , Genes, Tumor Suppressor , Genetic Heterogeneity , Humans , Iron/metabolism , Phosphoproteins/physiology , Protein Isoforms/genetics , RNA Splicing Factors , RNA, Messenger/genetics , Ribonucleoprotein, U2 Small Nuclear/physiology , Sequence Analysis, RNA , Signal Transduction/genetics
4.
FEBS J ; 281(23): 5194-207, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25238490

ABSTRACT

Alternative splicing of pre-mRNA, catalyzed by small nuclear ribonucleoproteins (snRNPs), plays an important role in proteome complexity and the modulation of cellular functions. snRNP polypeptide N (SmN), is tissue-specifically expressed, where it replaces snRNP polypeptide B (SmB)/B' in the Sm core assembly of snRNPs. Recent studies have demonstrated that perturbation of snRNPs leads to alternative splicing, but whether SmN modulates functions of the splicing machinery remains unclear. In this study, we found that ectopic expression of SmN increased utilization of the proximal 5' splice site on an adenovirus early gene 1A reporter. To evaluate the molecular mechanisms underlying SmN-dependent alternative splicing, we generated a HeLa cell line with an inducible expression system for SmN. Upon SmN induction, SmB/B' expression decreased dramatically, despite only small changes in the level and splicing pattern of SNRPB mRNA. In addition, SmN was incorporated into the U2 snRNP but not into the U1 snRNP after induction. Sedimentation analysis revealed a decrease in the level of mature U2 snRNP. This result suggests that SmN incorporation into the Sm core may impede processing, decreasing the level of functional U2 snRNP. We also found that the inclusion frequencies of alternatively spliced exons in the bridging integrator 1 and exocyst complex component 7 (EXOC7) genes were modulated by SmN expression. An enhanced GFP-EXOC7 reporter was used to confirm that SmN increases the inclusion frequency of EXOC7 exon 7. Taken together, our findings indicate that SmN expression reduces the level of mature U2 snRNP, leading to alternative splicing.


Subject(s)
Alternative Splicing , snRNP Core Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Cells, Cultured , Doxycycline/pharmacology , Fluorescent Antibody Technique , HeLa Cells , Humans , Nuclear Proteins/genetics , Ribonucleoprotein, U2 Small Nuclear/analysis , Ribonucleoprotein, U2 Small Nuclear/physiology , Tumor Suppressor Proteins/genetics , Vesicular Transport Proteins/genetics
5.
Mech Ageing Dev ; 135: 50-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24463145

ABSTRACT

Ageing in man is associated with changes to the splicing factor pool. A proportion of splicing factors are regulated during ageing by mechanisms involving the Ataxia Telangiectasia Mutated (ATM) gene, but the factors that determine the remaining proportion have yet to be identified. DNA methylation is known to be an important regulatory mechanism of gene expression. We assessed age-associated methylation and expression levels for 27 splicing factor genes, in peripheral blood samples from the InCHIANTI study. Examination of splicing patterns at specific loci was examined in a second cohort, the Exeter 10000 study. 27/502 methylation probes in 17 different genes were associated with age. Most changes were not associated with transcript expression levels or splicing patterns, but hypomethylation of the SF3B1 promoter region was found to mediate 53% of the relationship between age and transcript expression at this locus (p=0.02). DNA methylation does not appear to play a major role in regulation of the splicing factors, but changes in SF3B1 expression may be attributable to promoter hypomethylation at this locus. SF3B1 encodes a critical component of the U2 snRNP; altered expression of this gene may therefore contribute to the loss of regulated mRNA splicing that occurs with age.


Subject(s)
Aging , DNA Methylation , Gene Expression Regulation , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Cohort Studies , DNA , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Phosphoproteins/physiology , Promoter Regions, Genetic , RNA Splicing , RNA Splicing Factors , RNA, Messenger/metabolism , Ribonucleoprotein, U2 Small Nuclear/physiology , Young Adult
6.
PLoS One ; 7(10): e47621, 2012.
Article in English | MEDLINE | ID: mdl-23118884

ABSTRACT

The yeast Saccharomyces cerevisiae normally selects bud sites (and hence axes of cell polarization) in one of two distinct patterns, the axial pattern of haploid cells and the bipolar pattern of diploid cells. Although many of the proteins involved in bud-site selection are known, it is likely that others remain to be identified. Confirming a previous report (Ni and Snyder, 2001, Mol. Biol. Cell 12, 2147-2170), we found that diploids homozygous for deletions of IST3/SNU17 or BUD13 do not show normal bipolar budding. However, these abnormalities do not reflect defects in the apparatus of bipolar budding. Instead, the absence of Ist3 or Bud13 results in a specific defect in the splicing of the MATa1 pre-mRNA, which encodes a repressor that normally blocks expression of haploid-specific genes in diploid cells. When Mata1 protein is lacking, Axl1, a haploid-specific protein critical for the choice between axial and bipolar budding, is expressed ectopically in diploid cells and disrupts bipolar budding. The involvement of Ist3 and Bud13 in pre-mRNA splicing is by now well known, but the degree of specificity shown here for MATa1 pre-mRNA, which has no obvious basis in the pre-mRNA structure, is rather surprising in view of current models for the functions of these proteins. Moreover, we found that deletion of PML1, whose product is thought to function together with Ist3 and Bud13 in a three-protein retention-and-splicing (RES) complex, had no detectable effect on the splicing in vivo of either MATa1 or four other pre-mRNAs.


Subject(s)
Carrier Proteins , Homeodomain Proteins , RNA Splicing/genetics , Repressor Proteins , Ribonucleoprotein, U2 Small Nuclear , Saccharomyces cerevisiae Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Polarity/genetics , Cell Polarity/physiology , Diploidy , Haploidy , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology
8.
Blood ; 120(16): 3173-86, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22826563

ABSTRACT

Whole exome/genome sequencing has been fundamental in the identification of somatic mutations in the spliceosome machinery in myelodysplastic syndromes (MDSs) and other hematologic disorders. SF3B1, splicing factor 3b subunit 1 is mutated in 60%-80% of refractory anemia with ring sideroblasts (RARS) and RARS associated with thrombocytosis (RARS-T), 2 distinct subtypes of MDS and MDS/myeloproliferative neoplasms (MDSs/MPNs). An idiosyncratic feature of RARS/RARS-T is the presence of abnormal sideroblasts characterized by iron overload in the mitochondria, called RS. Based on the high frequency of mutations of SF3B1 in RARS/RARS-T, we investigated the consequences of SF3B1 alterations. Ultrastructurally, SF3B1 mutants showed altered iron distribution characterized by coarse iron deposits compared with wild-type RARS patients by transmission electron microscopy. SF3B1 knockdown experiments in K562 cells resulted in down-regulation of U2-type intron-splicing by RT-PCR. RNA-sequencing analysis of SF3B1 mutants showed differentially used genes relevant in MDS pathogenesis, such as ASXL1, CBL, EZH, and RUNX families. A SF3B pharmacologic inhibitor, meayamycin, induced the formation of RS in healthy BM cells. Further, BM aspirates of Sf3b1 heterozygous knockout mice showed RS by Prussian blue. In conclusion, we report the first experimental evidence of the association between SF3B1 and RS phenotype. Our data suggest that SF3B1 haploinsufficiency leads to RS formation.


Subject(s)
Anemia, Sideroblastic/pathology , Biomarkers, Tumor/genetics , Haploinsufficiency , Mutation/genetics , Myelodysplastic Syndromes/pathology , Phosphoproteins/metabolism , Phosphoproteins/physiology , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/physiology , Adolescent , Adult , Aged , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/metabolism , Animals , Biomarkers, Tumor/metabolism , Cells, Cultured , Female , Gene Expression Profiling , Humans , K562 Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Phosphoproteins/genetics , RNA Splicing Factors , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoprotein, U2 Small Nuclear/genetics , Young Adult
9.
Dev Biol ; 354(2): 232-41, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21504747

ABSTRACT

In Caenorhabditis elegans, germ cells develop as spermatids in the larva and as oocytes in the adult. Such fundamentally different gametes are produced through a fine-tuned balance between feminizing and masculinizing genes. For example, the switch to oogenesis requires repression of the fem-3 mRNA through the mog genes. Here we report on the cloning and characterization of the sex determination gene mog-2. MOG-2 is the worm homolog of spliceosomal protein U2A'. We found that MOG-2 is expressed in most nuclei of somatic and germ cells. In addition to its role in sex determination, mog-2 is required for meiosis. Moreover, MOG-2 binds to U2B″/RNP-3 in the absence of RNA. We also show that MOG-2 associates with the U2 snRNA in the absence of RNP-3. Therefore, we propose that MOG-2 is a bona fide component of the U2 snRNP. Albeit not being required for general pre-mRNA splicing, MOG-2 increases the splicing efficiency to a cryptic splice site that is located at the 5' end of the exon.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Meiosis , RNA Splice Sites , Ribonucleoprotein, U2 Small Nuclear/physiology , Sex Determination Processes , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Female , Germ Cells/physiology , Male , Molecular Sequence Data , Protein Binding , Ribonucleoprotein, U2 Small Nuclear/genetics
10.
PLoS One ; 6(1): e16077, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21283673

ABSTRACT

There is mounting evidence to suggest that the synthesis of pre-mRNA transcripts and their subsequent splicing are coordinated events. Previous studies have implicated the mammalian spliceosomal U2 snRNP as having a novel role in stimulating transcriptional elongation in vitro through interactions with the elongation factors P-TEFb and Tat-SF1; however, the mechanism remains unknown [1]. These factors are conserved in Saccharomyces cerevisiae, a fact that suggests that a similar interaction may occur in yeast to stimulate transcriptional elongation in vivo. To address this possibility we have looked for evidence of a role for the yeast Tat-SF1 homolog, Cus2, and the U2 snRNA in regulating transcription. Specifically, we have performed a genetic analysis to look for functional interactions between Cus2 or U2 snRNA and the P-TEFb yeast homologs, the Bur1/2 and Ctk1/2/3 complexes. In addition, we have analyzed Cus2-deleted or -overexpressing cells and U2 snRNA mutant cells to determine if they show transcription-related phenotypes similar to those displayed by the P-TEFb homolog mutants. In no case have we been able to observe phenotypes consistent with a role for either spliceosomal factor in transcription elongation. Furthermore, we did not find evidence for physical interactions between the yeast U2 snRNP factors and the P-TEFb homologs. These results suggest that in vivo, S. cerevisiae do not exhibit functional or physical interactions similar to those exhibited by their mammalian counterparts in vitro. The significance of the difference between our in vivo findings and the previously published in vitro results remains unclear; however, we discuss the potential importance of other factors, including viral proteins, in mediating the mammalian interactions.


Subject(s)
Gene Expression Regulation, Fungal , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes , Transcription, Genetic , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Protein Binding , Protein Kinases/genetics , RNA-Binding Proteins/genetics
11.
Bull Exp Biol Med ; 147(6): 733-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19902070

ABSTRACT

Intensive exercise triggers the cascade processes of body adaptation, including modulation of splisosome functioning, and can lead to modification of its activity and choice of alternative exons. We studied the effect of exercise of the maximum aerobic power on activation of transcription of genes involved in the splicing process. Short-term exercise resulted in a significant increase of mRNA expression of genes encoding proteins involved in the formation of precatalytic splisosome: DDX17, DDX46, HNRNPR, PRPF4B, and SRPK2. The role of the detected regulators in initiation of splisosome assembly under conditions of maximally intensive exercise is discussed.


Subject(s)
Exercise/physiology , Gene Expression Regulation , RNA Splicing/genetics , Adolescent , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/physiology , Gene Expression Profiling , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Humans , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/physiology , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/physiology , Young Adult
12.
FEMS Yeast Res ; 8(2): 276-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17995956

ABSTRACT

Using the Saccharomyces cerevisiae MATa/MATalpha ORF deletion collection, homozygous deletion strains were identified that undergo mating with MATa or MATalpha haploids. Seven homozygous deletions were identified that confer enhanced mating. Three of these, lacking CTF8, CTF18, and DCC1, mate at a low frequency with either MATa or MATalpha haploids. The products of these genes form a complex involved in sister chromatid cohesion. Each of these strains also exhibits increased chromosome loss rates, and mating likely occurs due to loss of one copy of chromosome III, which bears the MAT locus. Three other homozygous diploid deletion strains, ylr193cDelta/ylr193cDelta, yor305wDelta/yor305wDelta, and ypr170cDelta/ypr170cDelta, mate at very low frequencies with haploids of either or both mating types. However, an ist3Delta/ist3Delta strain mates only with MATa haploids. It is shown that IST3, previously linked to splicing, is required for efficient processing of the MATa1 message, particularly the first intron. As a result, the ist3Delta/ist3Delta strain expresses unbalanced ratios of Matalpha to Mata proteins and therefore mates with MATa haploids. Accordingly, mating in this diploid can be repressed by introduction of a MATa1 cDNA. In summary, this study underscores and elaborates upon predicted pathways by which mutations restore mating function to yeast diploids and identifies new mutants warranting further study.


Subject(s)
Genes, Mating Type, Fungal , Saccharomyces cerevisiae/physiology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/physiology , Chromosomes, Fungal/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Diploidy , Gene Deletion , Genes, Mating Type, Fungal/genetics , Genes, Mating Type, Fungal/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Sister Chromatid Exchange/genetics , Sister Chromatid Exchange/physiology
13.
PLoS Genet ; 2(12): e178, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17154718

ABSTRACT

The sequence of the SPF45 protein is significantly conserved, yet functional studies have identified it as a splicing factor in animal cells and as a DNA-repair protein in plants. Using a combined genetic and biochemical approach to investigate this apparent functional discrepancy, we unify and validate both of these studies by demonstrating that the Drosophila melanogaster protein is bifunctional, with independent functions in DNA repair and splicing. We find that SPF45 associates with the U2 snRNP and that mutations that remove the C-terminal end of the protein disrupt this interaction. Although animals carrying this mutation are viable, they are nevertheless compromised in their ability to regulate Sex-lethal splicing, demonstrating that Sex-lethal is an important physiological target of SPF45. Furthermore, these mutant animals exhibit phenotypes diagnostic of difficulties in recovering from exogenously induced DNA damage. The conclusion that SPF45 functions in the DNA-repair pathway is strengthened by finding both genetic and physical interactions between SPF45 and RAD201, a previously uncharacterized member of the RecA/Rad51 protein family. Together with our finding that the fly SPF45 protein increases the survival rate of mutagen-treated bacteria lacking the RecG helicase, these studies provide the tantalizing suggestion that SPF45 has an ancient and evolutionarily conserved role in DNA repair.


Subject(s)
DNA Repair/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , RNA Splicing , Ribonucleoprotein, U2 Small Nuclear/physiology , Amino Acid Sequence , Animals , DNA/genetics , DNA/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Exons , Heterochromatin/metabolism , Models, Genetic , Molecular Sequence Data , RNA Splicing Factors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism
14.
Mol Biol Cell ; 16(3): 1366-77, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15647371

ABSTRACT

The three subunits of human splicing factor SF3a are essential for the formation of the functional 17S U2 snRNP and prespliceosome assembly in vitro. RNAi-mediated depletion indicates that each subunit is essential for viability of human cells. Knockdown of single subunits results in a general block in splicing strongly suggesting that SF3a is a constitutive splicing factor in vivo. In contrast, splicing of several endogenous and reporter pre-mRNAs is not affected after knockdown of SF1, which functions at the onset of spliceosome assembly in vitro and is essential for cell viability. Thus, SF1 may only be required for the splicing of a subset of pre-mRNAs. We also observe a reorganization of U2 snRNP components in SF3a-depleted cells, where U2 snRNA and U2-B'' are significantly reduced in nuclear speckles and the nucleoplasm, but still present in Cajal bodies. Together with the observation that the 17S U2 snRNP cannot be detected in extracts from SF3a-depleted cells, our results provide further evidence for a function of Cajal bodies in U2 snRNP biogenesis.


Subject(s)
DNA-Binding Proteins/physiology , RNA Splicing , RNA, Messenger/metabolism , Ribonucleoprotein, U2 Small Nuclear/physiology , Transcription Factors/physiology , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , Cell Survival , Coiled Bodies/metabolism , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , RNA Interference , RNA Splicing Factors , RNA, Small Interfering/metabolism , RNA, Small Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/chemistry , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transfection
15.
Biochem J ; 382(Pt 1): 223-30, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15142036

ABSTRACT

SF3a (splicing factor 3a) complex is an essential component of U2 snRNPs (small nuclear ribonucleoprotein particles), which are involved in pre-mRNA splicing. This complex consists of three subunits: SF3a60, SF3a66 and SF3a120. Here, we report a possible non-canonical function of a well-characterized RNA-splicing factor, SF3a66. Ectopic expression experiments using each SF3a subunit in N1E 115 neuroblastoma cells reveals that SF3a66 alone can induce neurite extension, suggesting that SF3a66 functions in the regulation of cell morphology. A screen for proteins that bind to SF3a66 clarifies that SF3a66 binds to beta-tubulin, and also to microtubules, with high affinity, indicating that SF3a66 is a novel MAP (microtubule-associated protein). Electron microscopy experiments show that SF3a66 can bundle microtubules, and that bundling of microtubules is due to cross-bridging of microtubules by high-molecular-mass complexes of oligomerized SF3a66. These results indicate that SF3a66 is likely to be a novel MAP, and can function as a microtubule-bundling protein independently of RNA splicing.


Subject(s)
Microtubule-Associated Proteins/physiology , RNA Precursors/genetics , Ribonucleoprotein, U2 Small Nuclear/physiology , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Mice , Microtubules/metabolism , Neurites/metabolism , Neuroblastoma/chemistry , Neuroblastoma/pathology , Protein Binding/physiology , Protein Subunits/physiology , RNA Splicing/physiology , Ribonucleoproteins, Small Nuclear , Tubulin/metabolism
16.
Nucleic Acids Res ; 32(3): 1242-50, 2004.
Article in English | MEDLINE | ID: mdl-14973223

ABSTRACT

Mer1p activates the splicing of at least three pre-mRNAs (AMA1, MER2, MER3) during meiosis in the yeast Saccharomyces cerevisiae. We demonstrate that enhancer recognition by Mer1p is separable from Mer1p splicing activation. The C-terminal KH-type RNA-binding domain of Mer1p recognizes introns that contain the Mer1p splicing enhancer, while the N-terminal domain interacts with the spliceosome and activates splicing. Prior studies have implicated the U1 snRNP and recognition of the 5' splice site as key elements in Mer1p-activated splicing. We provide new evidence that Mer1p may also function at later steps of spliceosome assembly. First, Mer1p can activate splicing of introns that have mutated branch point sequences. Secondly, Mer1p fails to activate splicing in the absence of the non-essential U2 snRNP protein Snu17p. Thirdly, Mer1p interacts with the branch point binding proteins Mud2p and Bbp1p and the U2 snRNP protein Prp11p by two-hybrid assays. We conclude that Mer1p is a modular splicing regulator that can activate splicing at several early steps of spliceosome assembly and depends on the activities of both U1 and U2 snRNP proteins to activate splicing.


Subject(s)
RNA Splicing , RNA-Binding Proteins/physiology , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/physiology , Base Sequence , Introns , Mutation , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Spliceosomes/metabolism
17.
EMBO J ; 23(2): 376-85, 2004 Jan 28.
Article in English | MEDLINE | ID: mdl-14713954

ABSTRACT

Communication between U1 and U2 snRNPs is critical during pre-spliceosome assembly; yet, direct connections have not been observed. To investigate this assembly step, we focused on Prp5, an RNA-dependent ATPase of the DExD/H family. We identified homologs of Saccharomyces cerevisiae Prp5 in humans (hPrp5) and Schizosaccharomyces pombe (SpPrp5), and investigated their interactions and function. Depletion and reconstitution of SpPrp5 from extracts demonstrate that ATP binding and hydrolysis by Prp5 are required for pre-spliceosome complex A formation. hPrp5 and SpPrp5 are each physically associated with both U1 and U2 snRNPs; Prp5 contains distinct U1- and U2-interacting domains that are required for pre-spliceosome assembly; and, we observe a Prp5-associated U1/U2 complex in S. pombe. Together, these data are consistent with Prp5 being a bridge between U1 and U2 snRNPs at the time of pre-spliceosome formation.


Subject(s)
Adenosine Triphosphatases/physiology , Introns , RNA Helicases/physiology , RNA, Messenger/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Schizosaccharomyces pombe Proteins/physiology , Spliceosomes/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , DEAD-box RNA Helicases , Exons , Models, Genetic , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Precursors/metabolism , RNA, Messenger/chemistry , Ribonucleoprotein, U1 Small Nuclear/physiology , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Sequence Homology, Amino Acid
18.
Yeast ; 19(3): 193-202, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11816027

ABSTRACT

Cell tolerance to salt stress depends on many physiological functions, including the best characterized of osmotic adjustment, ion transport and sodium-sensitive sulphate metabolism. From a screening designed to identify novel determinants of salt tolerance we have isolated the YNL091w gene, probably an Ascomycete-specific gene encoding a protein of unknown function. This gene negatively affects salt tolerance and therefore has been designated NST1. The salt tolerance mechanism of nst1 mutants is novel because it is not related to osmoregulation, altered cation accumulation or sulphate metabolism. Genome-wide two-hybrid analysis has suggested that Nst1p interacts with the splicing factor Msl1p and, accordingly, the impact of NST1 on salt tolerance is dependent on a functional MSL1 gene. Loss of MSL1 and NST1 function has pleiotropic phenotypes including increased sensitivity to divalent cations (manganese and zinc) and to caffeine (a cell wall-weakening agent). On the other hand, msl1 mutants but not nst1 mutants are sensitive to thiabendazole (a microtubule-destabilizing agent) and to osmotic stress.


Subject(s)
Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/physiology , Sodium Chloride/pharmacology , Amino Acid Sequence , Autoantigens , Blotting, Northern , Caffeine/pharmacology , Lithium/metabolism , Molecular Sequence Data , Mutagenesis, Insertional , Phosphodiesterase Inhibitors/pharmacology , Potassium/analysis , RNA, Fungal/chemistry , RNA, Fungal/isolation & purification , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Sodium Chloride/metabolism , Thiabendazole/pharmacology , Two-Hybrid System Techniques , snRNP Core Proteins
19.
Nucleic Acids Res ; 29(19): 4006-13, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11574683

ABSTRACT

U12-dependent introns are found in small numbers in most eukaryotic genomes, but their scarcity makes accurate characterisation of their properties challenging. A computational search for U12-dependent introns was performed using the draft version of the human genome sequence. Human expressed sequences confirmed 404 U12-dependent introns within the human genome, a 6-fold increase over the total number of non-redundant U12-dependent introns previously identified in all genomes. Although most of these introns had AT-AC or GT-AG terminal dinucleotides, small numbers of introns with a surprising diversity of termini were found, suggesting that many of the non-canonical introns found in the human genome may be variants of U12-dependent introns and, thus, spliced by the minor spliceosome. Comparisons with U2-dependent introns revealed that the U12-dependent intron set lacks the 'short intron' peak characteristic of U2-dependent introns. Analysis of this U12-dependent intron set confirmed reports of a biased distribution of U12-dependent introns in the genome and allowed the identification of several alternative splicing events as well as a surprising number of apparent splicing errors. This new larger reference set of U12-dependent introns will serve as a resource for future studies of both the properties and evolution of the U12 spliceosome.


Subject(s)
Computational Biology/methods , Genome, Human , Introns , Ribonucleoproteins, Small Nuclear/physiology , Alternative Splicing , Humans , RNA Splice Sites , Ribonucleoprotein, U2 Small Nuclear/physiology
20.
Mol Cell Biol ; 21(9): 3037-46, 2001 May.
Article in English | MEDLINE | ID: mdl-11287609

ABSTRACT

We have isolated and microsequenced Snu17p, a novel yeast protein with a predicted molecular mass of 17 kDa that contains an RNA recognition motif. We demonstrate that Snu17p binds specifically to the U2 small nuclear ribonucleoprotein (snRNP) and that it is part of the spliceosome, since the pre-mRNA and the lariat-exon 2 are specifically coprecipitated with Snu17p. Although the SNU17 gene is not essential, its knockout leads to a slow-growth phenotype and to a pre-mRNA splicing defect in vivo. In addition, the first step of splicing is dramatically decreased in extracts prepared from the snu17 deletion (snu17Delta) mutant. This defect is efficiently reversed by the addition of recombinant Snu17p. To investigate the step of spliceosome assembly at which Snu17p acts, we have used nondenaturing gel electrophoresis. In Snu17p-deficient extracts, the spliceosome runs as a single slowly migrating complex. In wild-type extracts, usually at least two distinct complexes are observed: the prespliceosome, or B complex, containing the U2 but not the U1 snRNP, and the catalytically active spliceosome, or A complex, containing the U2, U6, and U5 snRNPs. Northern blot analysis and affinity purification of the snu17Delta spliceosome showed that it contains the U1, U2, U6, U5, and U4 snRNPs. The unexpected stabilization of the U1 snRNP and the lack of dissociation of the U4 snRNP suggest that loss of Snu17p inhibits the progression of spliceosome assembly prior to U1 snRNP release and after [U4/U6.U5] tri-snRNP addition.


Subject(s)
Fungal Proteins/metabolism , RNA Splicing , Ribonucleoprotein, U2 Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins , Spliceosomes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Catalysis , DNA, Fungal , Fungal Proteins/genetics , Fungal Proteins/physiology , Humans , Molecular Sequence Data , Mutagenesis , Phenotype , RNA Precursors , RNA, Fungal/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Spliceosomes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...