Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33876749

ABSTRACT

Most genes associated with acute myeloid leukemia (AML) are mutated in less than 10% of patients, suggesting that alternative mechanisms of gene disruption contribute to this disease. Here, we find a set of splicing events that alter the expression of a subset of AML-associated genes independent of known somatic mutations. In particular, aberrant splicing triples the number of patients with reduced functional EZH2 compared with that predicted by somatic mutation alone. In addition, we unexpectedly find that the nonsense-mediated decay factor DHX34 exhibits widespread alternative splicing in sporadic AML, resulting in a premature stop codon that phenocopies the loss-of-function germline mutations observed in familial AML. Together, these results demonstrate that classical mutation analysis underestimates the burden of functional gene disruption in AML and highlight the importance of assessing the contribution of alternative splicing to gene dysregulation in human disease.


Subject(s)
Alternative Splicing , Leukemia, Myeloid, Acute/genetics , Mutation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Genotype , Humans , Nonsense Mediated mRNA Decay , RNA Helicases/genetics , RNA Helicases/metabolism
2.
Cell Chem Biol ; 28(8): 1145-1157.e6, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33689684

ABSTRACT

Dysregulated pre-mRNA splicing is an emerging Achilles heel of cancers and myelodysplasias. To expand the currently limited portfolio of small-molecule drug leads, we screened for chemical modulators of the U2AF complex, which nucleates spliceosome assembly and is mutated in myelodysplasias. A hit compound specifically enhances RNA binding by a U2AF2 subunit. Remarkably, the compound inhibits splicing of representative substrates and stalls spliceosome assembly at the stage of U2AF function. Computational docking, together with structure-guided mutagenesis, indicates that the compound bridges the tandem U2AF2 RNA recognition motifs via hydrophobic and electrostatic moieties. Cells expressing a cancer-associated U2AF1 mutant are preferentially killed by treatment with the compound. Altogether, our results highlight the potential of trapping early spliceosome assembly as an effective pharmacological means to manipulate pre-mRNA splicing. By extension, we suggest that stabilizing assembly intermediates may offer a useful approach for small-molecule inhibition of macromolecular machines.


Subject(s)
RNA Precursors/drug effects , RNA Splicing/drug effects , RNA, Neoplasm/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Splicing Factor U2AF/antagonists & inhibitors , Female , HEK293 Cells , Humans , K562 Cells , Molecular Docking Simulation , Molecular Structure , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
3.
Nucleic Acids Res ; 48(10): 5710-5719, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32338744

ABSTRACT

RNA binding proteins (RBPs) frequently regulate the expression of other RBPs in mammalian cells. Such cross-regulation has been proposed to be important to control networks of coordinated gene expression; however, much remains to be understood about how such networks of cross-regulation are established and what the functional consequence is of coordinated or reciprocal expression of RBPs. Here we demonstrate that the RBPs CELF2 and hnRNP C regulate the expression of each other, such that depletion of one results in reduced expression of the other. Specifically, we show that loss of hnRNP C reduces the transcription of CELF2 mRNA, while loss of CELF2 results in decreased efficiency of hnRNP C translation. We further demonstrate that this reciprocal regulation serves to fine tune the splicing patterns of many downstream target genes. Together, this work reveals new activities of hnRNP C and CELF2, provides insight into a previously unrecognized gene regulatory network, and demonstrates how cross-regulation of RBPs functions to shape the cellular transcriptome.


Subject(s)
CELF Proteins/metabolism , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Nerve Tissue Proteins/metabolism , Protein Biosynthesis , RNA Splicing , Transcription, Genetic , CELF Proteins/biosynthesis , CELF Proteins/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/biosynthesis , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Humans , Jurkat Cells , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , T-Lymphocytes/metabolism
4.
Cell Rep ; 28(11): 2795-2806.e3, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31509743

ABSTRACT

The 3' UTR (UTR) of human mRNAs plays a critical role in controlling protein expression and function. Importantly, 3' UTRs of human messages are not invariant for each gene but rather are shaped by alternative polyadenylation (APA) in a cell state-dependent manner, including in response to T cell activation. However, the proteins and mechanisms driving APA regulation remain poorly understood. Here we show that the RNA-binding protein CELF2 controls APA of its own message in a signal-dependent manner by competing with core enhancers of the polyadenylation machinery for binding to RNA. We further show that CELF2 binding overlaps with APA enhancers transcriptome-wide, and almost half of 3' UTRs that undergo T cell signaling-induced APA are regulated in a CELF2-dependent manner. These studies thus reveal CELF2 to be a critical regulator of 3' UTR identity in T cells and demonstrate an additional mechanism for CELF2 in regulating polyadenylation site choice.


Subject(s)
CELF Proteins/metabolism , Gene Expression Regulation/genetics , Nerve Tissue Proteins/metabolism , Polyadenylation/genetics , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , CELF Proteins/genetics , Cell Line, Tumor , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Enhancer Elements, Genetic , Humans , Introns/genetics , Nerve Tissue Proteins/genetics , Protein Binding , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Seq , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism , Transcriptome
5.
Biophys J ; 111(12): 2570-2586, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-28002734

ABSTRACT

Splicing factor 1 (SF1) recognizes 3' splice sites of the major class of introns as a ternary complex with U2AF65 and U2AF35 splicing factors. A conserved SPSP motif in a coiled-coil domain of SF1 is highly phosphorylated in proliferating human cells and is required for cell proliferation. The UHM kinase 1 (UHMK1), also called KIS, double-phosphorylates both serines of this SF1 motif. Here, we use isothermal titration calorimetry to demonstrate that UHMK1 phosphorylation of the SF1 SPSP motif slightly enhances specific binding of phospho-SF1 to its cognate U2AF65 protein partner. Conversely, quantitative fluorescence anisotropy RNA binding assays and isothermal titration calorimetry experiments establish that double-SPSP phosphorylation reduces phospho-SF1 and phospho-SF1-U2AF65 binding affinities for either optimal or suboptimal splice-site RNAs. Domain-substitution and mutagenesis experiments further demonstrate that arginines surrounding the phosphorylated SF1 loop are required for cooperative 3' splice site recognition by the SF1-U2AF65 complex (where cooperativity is defined as a nonadditive increase in RNA binding by the protein complex relative to the individual proteins). In the context of local, intracellular concentrations, the subtle effects of SF1 phosphorylation on its associations with U2AF65 and splice-site RNAs are likely to influence pre-mRNA splicing. However, considering roles for SF1 in pre-mRNA retention and transcriptional repression, as well as in splicing, future comprehensive investigations are needed to fully explain the requirement for SF1 SPSP phosphorylation in proliferating human cells.


Subject(s)
RNA Splice Sites , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Base Sequence , Humans , Models, Molecular , Mutation , Phosphorylation , Protein Domains , RNA Splice Sites/genetics , RNA Splicing Factors/genetics
6.
PLoS Genet ; 12(10): e1006384, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27776121

ABSTRACT

We have asked how the common S34F mutation in the splicing factor U2AF1 regulates alternative splicing in lung cancer, and why wild-type U2AF1 is retained in cancers with this mutation. A human lung epithelial cell line was genetically modified so that U2AF1S34F is expressed from one of the two endogenous U2AF1 loci. By altering levels of mutant or wild-type U2AF1 in this cell line and by analyzing published data on human lung adenocarcinomas, we show that S34F-associated changes in alternative splicing are proportional to the ratio of S34F:wild-type gene products and not to absolute levels of either the mutant or wild-type factor. Preferential recognition of specific 3' splice sites in S34F-expressing cells is largely explained by differential in vitro RNA-binding affinities of mutant versus wild-type U2AF1 for those same 3' splice sites. Finally, we show that lung adenocarcinoma cell lines bearing U2AF1 mutations do not require the mutant protein for growth in vitro or in vivo. In contrast, wild-type U2AF1 is required for survival, regardless of whether cells carry the U2AF1S34F allele. Our results provide mechanistic explanations of the magnitude of splicing changes observed in U2AF1-mutant cells and why tumors harboring U2AF1 mutations always retain an expressed copy of the wild-type allele.


Subject(s)
Adenocarcinoma/genetics , Alternative Splicing/genetics , Cell Proliferation/genetics , Lung Neoplasms/genetics , Splicing Factor U2AF/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice , Mutant Proteins/genetics , Mutation , Nucleotide Motifs/genetics , RNA, Messenger/biosynthesis , Splicing Factor U2AF/biosynthesis , Transcriptome/genetics , Xenograft Model Antitumor Assays
7.
Nat Commun ; 7: 10950, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26952537

ABSTRACT

How the essential pre-mRNA splicing factor U2AF(65) recognizes the polypyrimidine (Py) signals of the major class of 3' splice sites in human gene transcripts remains incompletely understood. We determined four structures of an extended U2AF(65)-RNA-binding domain bound to Py-tract oligonucleotides at resolutions between 2.0 and 1.5 Å. These structures together with RNA binding and splicing assays reveal unforeseen roles for U2AF(65) inter-domain residues in recognizing a contiguous, nine-nucleotide Py tract. The U2AF(65) linker residues between the dual RNA recognition motifs (RRMs) recognize the central nucleotide, whereas the N- and C-terminal RRM extensions recognize the 3' terminus and third nucleotide. Single-molecule FRET experiments suggest that conformational selection and induced fit of the U2AF(65) RRMs are complementary mechanisms for Py-tract association. Altogether, these results advance the mechanistic understanding of molecular recognition for a major class of splice site signals.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Splice Sites , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Crystallography, X-Ray , Humans , Nuclear Proteins/genetics , Protein Structure, Tertiary , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , Ribonucleoproteins/genetics , Splicing Factor U2AF
SELECTION OF CITATIONS
SEARCH DETAIL
...