scholarly journals U2AF1 mutations alter splice site recognition in hematological malignancies

2013 ◽  
Author(s):  
Janine O Ilagan ◽  
Aravind Ramakrishnan ◽  
Brian Hayes ◽  
Michele E Murphy ◽  
Ahmad S Zebari ◽  
...  

Whole-exome sequencing studies have identified common mutations affecting genes encoding components of the RNA splicing machinery in hematological malignancies. Here, we sought to determine how mutations affecting the 3' splice site recognition factor U2AF1 alter its normal role in RNA splicing. We find that U2AF1 mutations influence the similarity of splicing programs in leukemias, but do not give rise to widespread splicing failure. U2AF1 mutations cause differential splicing of hundreds of genes, affecting biological pathways such as DNA methylation (DNMT3B), X chromosome inactivation (H2AFY), the DNA damage response (ATR, FANCA), and apoptosis (CASP8). We show that U2AF1 mutations alter the preferred 3' splice site motif in patients, in cell culture, and in vitro. Mutations affecting the first and second zinc fingers give rise to different alterations in splice site preference and largely distinct downstream splicing programs. These allele-specific effects are consistent with a computationally predicted model of U2AF1 in complex with RNA. Our findings suggest that U2AF1 mutations contribute to pathogenesis by causing quantitative changes in splicing that affect diverse cellular pathways, and give insight into the normal function of U2AF1's zinc finger domains.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1889-1889 ◽  
Author(s):  
Janine O. Ilagan ◽  
Aravind Ramakrishnan ◽  
Brian Hayes ◽  
Michele E. Murphy ◽  
Ahmad S. Zebari ◽  
...  

Abstract Mutations affecting the spliceosomal protein U2AF1 are among the most common mutations observed in patients with MDS and related disorders. However, it is unclear how these mutations affect the normal RNA splicing process, and how the resulting changes in splicing contribute to myeloid dysplasia. Here, we combined the strengths of data from primary AML patient samples with the controlled context of isogenic cell lines. We generated K562 erythroleukemic cell lines stably expressing each of the four common U2AF1 mutations (S34F, S34Y, Q157P, and Q157R). We compared expression of each of these mutant alleles with knock down of endogenous U2AF1 to compare the relative consequences of U2AF1 mutations versus loss of function. We first sought to identify changes in splicing driven by U2AF1 mutations that contribute to myeloid dysplasia. We compared the splicing of ~125,000 annotated alternative splicing events and ~160,000 constitutively spliced junctions between AML samples with or without mutations (TCGA cohort), as well as our isogenic K562 cell lines stably expressing either mutant (S34F, S34Y, Q157P, and Q157R) or wild-type (WT) alleles of U2AF1. Unsupervised cluster analysis revealed that S34F/Y versus Q157P/R samples clustered together in both the AML data and our cell lines, suggesting that U2AF1 mutations affecting different residues of the protein have different molecular consequences. Intersecting the AML and K562 data, we identified >300 splicing events that were consistently differentially spliced in association with S34 mutations, and a similar number for Q157 mutations. Many of these splicing events affected biological pathways that have been implicated in myeloid malignancies, including DNA methylation (DNMT3B), X chromosome inactivation (H2AFY), the DNA damage response (ATR, FANCA), and apoptosis (CASP8). For example, two exons of DNMT3B are differentially spliced in both AML samples and our K562 cells (Figure A), including an exon lying within the methyltransferase domain. We next identified mechanistic changes in the splicing process caused by U2AF1 mutations. U2AF1 binds the intron-exon boundary by sequence-specifically recognizing the AG dinucleotide and flanking sequence positions that define the 3' splice site. Comparing AML samples and K562 cells with and without U2AF1 mutations, we found that S34 and Q157 mutations give rise to specific and distinct alterations in 3' splice site preference. S34 mutations alter the consensus nucleotide immediately before the AG dinucleotide, while Q157 mutations alter the consensus nucleotide immediately after the AG (Figure B). We observed highly similar allele-specific alterations in 3' splice site preference in every AML patient with a U2AF1 mutation, as well as all K562 cell lines expressing a U2AF1 mutant allele. In contrast, knock down of endogenous U2AF1 caused no alterations in the consensus sequence at those positions, indicating that U2AF1 mutations do not cause loss of function at the level of RNA splicing. To confirm that the nucleotides immediately before and after the AG determine whether a splice site responds to U2AF1 mutations, we created minigenes of cassette exons within the ATR and EPB49 genes. We found that response to U2AF1 S34 and Q157 mutations requires the endogenous nucleotides immediately before and after the AG, as predicted by our genomics analysis, and that mutating these positions abolishes response to U2AF1 mutations. Finally, we recapitulated the RNA splicing process in vitro using nuclear extract from blood cells expressing either wild-type or mutant U2AF1 to show that identical changes in splice site preference occur in a controlled in vitro context (Figure C). Together, our data show that U2AF1 mutations cause allele-specific alterations in normal 3' splice site recognition in patients, in isogenic cell lines, and in vitro. These alterations in splice site preference give rise to mis-splicing that affects many genes previously implicated in myeloid malignancies. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


1993 ◽  
Vol 13 (5) ◽  
pp. 2677-2687 ◽  
Author(s):  
D A Sterner ◽  
S M Berget

Very small vertebrate exons are problematic for RNA splicing because of the proximity of their 3' and 5' splice sites. In this study, we investigated the recognition of a constitutive 7-nucleotide mini-exon from the troponin I gene that resides quite close to the adjacent upstream exon. The mini-exon failed to be included in spliced RNA when placed in a heterologous gene unless accompanied by the upstream exon. The requirement for the upstream exon disappeared when the mini-exon was internally expanded, suggesting that the splice sites bordering the mini-exon are compatible with those of other constitutive vertebrate exons and that the small size of the exon impaired inclusion. Mutation of the 5' splice site of the natural upstream exon did not result in either exon skipping or activation of a cryptic 5' splice site, the normal vertebrate phenotypes for such mutants. Instead, a spliced RNA accumulated that still contained the upstream intron. In vitro, the mini-exon failed to assemble into spliceosome complexes unless either internally expanded or accompanied by the upstream exon. Thus, impaired usage of the mini-exon in vivo was accompanied by impaired recognition in vitro, and recognition of the mini-exon was facilitated by the presence of the upstream exon in vivo and in vitro. Cumulatively, the atypical in vivo and in vitro properties of the troponin exons suggest a mechanism for the recognition of this mini-exon in which initial recognition of an exon-intron-exon unit is followed by subsequent recognition of the intron.


1999 ◽  
Vol 73 (3) ◽  
pp. 2394-2400 ◽  
Author(s):  
Craig R. Cook ◽  
Mark T. McNally

ABSTRACT The negative regulator of splicing (NRS) from Rous sarcoma virus suppresses viral RNA splicing and is one of several ciselements that account for the accumulation of large amounts of unspliced RNA for use as gag-pol mRNA and progeny virion genomic RNA. The NRS can also inhibit splicing of heterologous introns in vivo and in vitro. Previous data showed that the splicing factors SF2/ASF and U1, U2, and U11 small nuclear ribonucleoproteins (snRNPs) bind the NRS, and a correlation was established between SF2/ASF and U11 binding and activity, suggesting that these factors are important for function. These observations, and the finding that a large spliceosome-like complex (NRS-C) assembles on NRS RNA in nuclear extract, led to the proposal that the NRS is recognized as a minor-class 5′ splice site. One model to explain NRS splicing inhibition holds that the NRS interacts nonproductively with and sequesters U2-dependent 3′ splice sites. In this study, we provide evidence that the NRS interacts with an adenovirus 3′ splice site. The interaction was dependent on the integrity of the branch point and pyrimidine tract of the 3′ splice site, and it was sensitive to a mutation that was previously shown to abolish U11 snRNP binding and NRS function. However, further mutational analyses of NRS sequences have identified a U1 binding site that overlaps the U11 site, and the interaction with the 3′ splice site correlated with U1, not U11, binding. These results show that the NRS can interact with a 3′ splice site and suggest that U1 is of primary importance for NRS splicing inhibition.


1999 ◽  
Vol 13 (6) ◽  
pp. 740-753 ◽  
Author(s):  
E. Labourier ◽  
H.-M. Bourbon ◽  
I.-e. Gallouzi ◽  
M. Fostier ◽  
E. Allemand ◽  
...  

1993 ◽  
Vol 13 (5) ◽  
pp. 2677-2687
Author(s):  
D A Sterner ◽  
S M Berget

Very small vertebrate exons are problematic for RNA splicing because of the proximity of their 3' and 5' splice sites. In this study, we investigated the recognition of a constitutive 7-nucleotide mini-exon from the troponin I gene that resides quite close to the adjacent upstream exon. The mini-exon failed to be included in spliced RNA when placed in a heterologous gene unless accompanied by the upstream exon. The requirement for the upstream exon disappeared when the mini-exon was internally expanded, suggesting that the splice sites bordering the mini-exon are compatible with those of other constitutive vertebrate exons and that the small size of the exon impaired inclusion. Mutation of the 5' splice site of the natural upstream exon did not result in either exon skipping or activation of a cryptic 5' splice site, the normal vertebrate phenotypes for such mutants. Instead, a spliced RNA accumulated that still contained the upstream intron. In vitro, the mini-exon failed to assemble into spliceosome complexes unless either internally expanded or accompanied by the upstream exon. Thus, impaired usage of the mini-exon in vivo was accompanied by impaired recognition in vitro, and recognition of the mini-exon was facilitated by the presence of the upstream exon in vivo and in vitro. Cumulatively, the atypical in vivo and in vitro properties of the troponin exons suggest a mechanism for the recognition of this mini-exon in which initial recognition of an exon-intron-exon unit is followed by subsequent recognition of the intron.


Author(s):  
Noriko Fujihara ◽  
Kazuyoshi Yamauchi ◽  
Masako Hirota-Kawadobora ◽  
Shinsuke Ishikawa ◽  
Minoru Tozuka ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. SCI-14-SCI-14
Author(s):  
Seishi Ogawa

Abstract Abstract SCI-14 During the past decade, significant progress has been made in our understanding of the molecular pathogenesis of myelodysplastic syndromes (MDS) and related myeloid neoplasms, in which one of the major findings was frequent mutations of genes in epigenetic regulation, such as DNA methylation (DNMT3A, TET2, and IDH1/2) and chromatin modifications (ASXL1, EZH2, EED, and SUZ12). They are also found in comparable or even higher fractions of other myeloid neoplasms, underscoring the common impact of deregulated epigenetic regulation on myeloid leukemogenesis. On the other hand, a new class of pathway mutations has been uncovered recently that commonly involve the RNA splicing machinery (1, 2, 3). Thus, at least eight different components of the machinery, invariably engaged in the 3' splice site recognition, have been reported to be mutated in as high as 45 percent to 85 percent of cases with different subtypes of MDS and related myeloid neoplasms mostly in a mutually exclusive manner. This indicates that the 3' splice site recognition is the functional target of these mutations. There exist discrete mutational hotspots in three out of four major targets, including SF3B1, SRSF2, U2AF35, and ZRSR2, and these mutant alleles can induce abnormal RNA splicing, indicating the gain-of-function nature of the mutations (2). Splicing factor mutations are largely specific to myelodysplasia phenotypes but relatively rare in acute myeloid leukemia and myeloproliferative neoplasms (2), suggesting their primary roles in the pathogenesis of myelodysplasia. The genotype-phenotype association is especially prominent in the case of SF3B1 mutations, which were found in 76 percent to 83 percent of cases of refractory anemia with ringed sideroblasts (RARS), refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T), and refractory cytopenia with multilineage dysplasia and ringed sideroblasts (RCMD-RS) (1, 2). In this session, the updated findings on the spliceosome mutations found in myelodysplasia, including their clinical and functional aspects, will be discussed. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 25 (1) ◽  
pp. 14-26 ◽  
Author(s):  
Janine O. Ilagan ◽  
Aravind Ramakrishnan ◽  
Brian Hayes ◽  
Michele E. Murphy ◽  
Ahmad S. Zebari ◽  
...  

1993 ◽  
Vol 342 (1301) ◽  
pp. 217-224 ◽  

Pre-mRNA splicing or the removal of introns from precursor messenger RNAs depends on the accurate recognition of intron sequences by the plant splicing machinery. The major components of this machinery are small nuclear ribonucleoprotein protein particles (snRNPs) which consist of snRNAs and snRNP proteins. We have analysed various aspects of intron sequence and structure in relation to splice site selection and splicing efficiency and we have cloned snRNA genes and a gene encoding the snRNP protein, U2B". In the absence of an in vitro splicing system for plants, transient expression in protoplasts and stable plant transform ations have been used to analyse splicing of intron constructs. We aim to address the function of the UsnRNP-specific protein, U2B", via the production of transgenic plants expressing antisense U2B" transcripts and epitope-tagged U2B" protein. In addition, we have cloned genes encoding other proteins which potentially interact with RNA, such as RNA helicases, and strategies involving transgenic plants are being developed to analyse their function.


2018 ◽  
Author(s):  
Kaushik Saha ◽  
Mike Minh Fernandez ◽  
Tapan Biswas ◽  
Charles Leonard Mallari Lumba ◽  
Gourisankar Ghosh

ABSTRACTFor splicing of a metazoan pre-mRNA, the four major splice signals – 5′ and 3′ splice sites (SS), branch-point site (BS), and a poly-pyrimidine tract (PPT) – are initially bound by splicing factors U1 snRNP, U2AF35, SF1, and U2AF65, respectively, leading up to an early spliceosomal complex, the E-complex. The E-complex consists of additional components and the mechanism of its assembly is unclear. Hence, how splice signals are organized within E-complex defining the exon-intron boundaries remains elusive. Here we present in vitro stepwise reconstitution of an early spliceosome, assembled by cooperative actions of U1 snRNP, SRSF1, SF1, U2AF65, U2AF35, and hnRNP A1, termed here the recognition (R) complex, within which both splice sites are recognized. The R-complex assembly indicates that the SRSF1:pre-mRNA complex initially defines a substrate for U1 snRNP, engaging exons at both ends of an intron. Subsequent 5′SS-dependent U1 snRNP binding enables recognition of the remaining splice signals, defining the intron. This R-complex assembly indicates the minimal constituents for intron definition revealing mechanistic principles behind the splice site recognition.


Sign in / Sign up

Export Citation Format

Share Document