Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.083
Filter
1.
Article in English | MEDLINE | ID: mdl-38862431

ABSTRACT

Ribonuclease P (RNase P) was first described in the 1970's as an endoribonuclease acting in the maturation of precursor transfer RNAs (tRNAs). More recent studies, however, have uncovered non-canonical roles for RNase P and its components. Here, we review the recent progress of its involvement in chromatin assembly, DNA damage response, and maintenance of genome stability with implications in tumorigenesis. The possibility of RNase P as a therapeutic target in cancer is also discussed.


Subject(s)
Neoplasms , RNA Precursors , RNA, Transfer , Ribonuclease P , Ribonuclease P/metabolism , Ribonuclease P/genetics , Humans , RNA, Transfer/metabolism , RNA, Transfer/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/enzymology , RNA Precursors/metabolism , RNA Precursors/genetics , Genomic Instability , Animals , DNA Damage , RNA Processing, Post-Transcriptional , Chromatin Assembly and Disassembly/genetics
2.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891854

ABSTRACT

MicroRNAs (miRNAs) regulate approximately one-third of all human genes. The dysregulation of miRNAs has been implicated in the development of numerous human diseases, including cancers. In our investigation focusing on altering specific miRNA expression in human pancreatic cancer cells, we encountered an interesting finding. While two expression vector designs effectively enhanced miR-708 levels, they were unable to elevate mature forms of miR-29b, -1290, -2467, and -6831 in pancreatic cancer cell lines. This finding was also observed in a panel of other non-pancreatic cancer cell lines, suggesting that miRNA processing efficiency was cell line specific. Using a step-by-step approach in each step of miRNA processing, we ruled out alternative strand selection by the RISC complex and transcriptional interference at the primary miRNA (pri-miRNA) level. DROSHA processing and pri-miRNA export from the nucleus also appeared to be occurring normally. We observed precursor (pre-miRNA) accumulation only in cell lines where mature miRNA expression was not achieved, suggesting that the block was occurring at the pre-miRNA stage. To further confirm this, synthetic pre-miRNA mimics that bypass DICER processing were processed into mature miRNAs in all cases. This study has demonstrated the distinct behaviours of different miRNAs with the same vector in the same cell line, the same miRNA between the two vector designs, and with the same miRNA across different cell lines. We identified a stable vector pre-miRNA processing block. Our findings on the structural and sequence differences between successful and non-successful vector designs could help to inform future chimeric miRNA design strategies and act as a guide to other researchers on the intricate processing dynamics that can impact vector efficiency. Our research confirms the potential of miRNA mimics to surmount some of these complexities.


Subject(s)
MicroRNAs , Pancreatic Neoplasms , RNA Processing, Post-Transcriptional , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA Processing, Post-Transcriptional/genetics , Cell Line, Tumor , Ribonuclease III/metabolism , Ribonuclease III/genetics , Gene Expression Regulation, Neoplastic , Transfection , RNA Precursors/genetics , RNA Precursors/metabolism , Animals
3.
PLoS Genet ; 20(6): e1011316, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833506

ABSTRACT

Splicing is an important step of gene expression regulation in eukaryotes, as there are many mRNA precursors that can be alternatively spliced in different tissues, at different cell cycle phases or under different external stimuli. We have developed several integrated fluorescence-based in vivo splicing reporter constructs that allow the quantification of fission yeast splicing in vivo on intact cells, and we have compared their splicing efficiency in a wild type strain and in a prp2-1 (U2AF65) genetic background, showing a clear dependency between Prp2 and a consensus signal at 5' splicing site (5'SS). To isolate novel genes involved in regulated splicing, we have crossed the reporter showing more intron retention with the Schizosaccharomyces pombe knock out collection. Among the candidate genes involved in the regulation of splicing, we have detected strong splicing defects in two of the mutants -Δcwf12, a member of the NineTeen Complex (NTC) and Δsaf5, a methylosome subunit that acts together with the survival motor neuron (SMN) complex in small nuclear ribonucleoproteins (snRNP) biogenesis. We have identified that strains with mutations in cwf12 have inefficient splicing, mainly when the 5'SS differs from the consensus. However, although Δsaf5 cells also have some dependency on 5'SS sequence, we noticed that when one intron of a given pre-mRNA was affected, the rest of the introns of the same pre-mRNA had high probabilities of being also affected. This observation points Saf5 as a link between transcription rate and splicing.


Subject(s)
RNA Splicing , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Transcription, Genetic , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Gene Expression Regulation, Fungal , Introns/genetics , Mutation , Alternative Splicing/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splice Sites/genetics , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
4.
Proc Natl Acad Sci U S A ; 121(27): e2406710121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917004

ABSTRACT

The essential role of U4 snRNP in pre-messenger RNA (mRNA) splicing has been well established. In this study, we utilized an antisense morpholino oligonucleotide (AMO) specifically targeting U4 snRNA to achieve functional knockdown of U4 snRNP in HeLa cells. Our results showed that this knockdown resulted in global intronic premature cleavage and polyadenylation (PCPA) events, comparable to the effects observed with U1 AMO treatment, as demonstrated by mRNA 3'-seq analysis. Furthermore, our study suggested that this may be a common phenomenon in both human and mouse cell lines. Additionally, we showed that U4 AMO treatment disrupted transcription elongation, as evidenced by chromatin immunoprecipitation sequencing (ChIP-seq) analysis for RNAPII. Collectively, our results identified a unique role for U4 snRNP in the inhibition of PCPA and indicated a model wherein splicing intrinsically inhibits intronic cleavage and polyadenylation in the context of cotranscriptional mRNA processing.


Subject(s)
Polyadenylation , RNA Precursors , RNA Splicing , Humans , RNA Precursors/metabolism , RNA Precursors/genetics , HeLa Cells , Mice , Animals , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Introns/genetics
5.
Nat Commun ; 15(1): 4683, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824131

ABSTRACT

The human mitochondrial genome is transcribed into two RNAs, containing mRNAs, rRNAs and tRNAs, all dedicated to produce essential proteins of the respiratory chain. The precise excision of tRNAs by the mitochondrial endoribonucleases (mt-RNase), P and Z, releases all RNA species from the two RNA transcripts. The tRNAs then undergo 3'-CCA addition. In metazoan mitochondria, RNase P is a multi-enzyme assembly that comprises the endoribonuclease PRORP and a tRNA methyltransferase subcomplex. The requirement for this tRNA methyltransferase subcomplex for mt-RNase P cleavage activity, as well as the mechanisms of pre-tRNA 3'-cleavage and 3'-CCA addition, are still poorly understood. Here, we report cryo-EM structures that visualise four steps of mitochondrial tRNA maturation: 5' and 3' tRNA-end processing, methylation and 3'-CCA addition, and explain the defined sequential order of the tRNA processing steps. The methyltransferase subcomplex recognises the pre-tRNA in a distinct mode that can support tRNA-end processing and 3'-CCA addition, likely resulting from an evolutionary adaptation of mitochondrial tRNA maturation complexes to the structurally-fragile mitochondrial tRNAs. This subcomplex can also ensure a tRNA-folding quality-control checkpoint before the sequential docking of the maturation enzymes. Altogether, our study provides detailed molecular insight into RNA-transcript processing and tRNA maturation in human mitochondria.


Subject(s)
Mitochondria , RNA, Transfer , Ribonuclease P , tRNA Methyltransferases , Humans , RNA, Transfer/metabolism , RNA, Transfer/genetics , RNA, Transfer/chemistry , Mitochondria/metabolism , Ribonuclease P/metabolism , Ribonuclease P/genetics , Ribonuclease P/chemistry , tRNA Methyltransferases/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/chemistry , RNA Processing, Post-Transcriptional , Cryoelectron Microscopy , RNA, Mitochondrial/metabolism , RNA, Mitochondrial/genetics , RNA, Mitochondrial/chemistry , Methylation , Nucleic Acid Conformation , Models, Molecular , RNA Precursors/metabolism , RNA Precursors/genetics
6.
Biochem Pharmacol ; 225: 116251, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38701867

ABSTRACT

Hepatocellular carcinoma (HCC) is the main histological subtype of primary liver cancer and remains one of the most common solid malignancies globally. Ferroptosis was recently defined as an iron-catalyzed form of regulated necrosis. Because cancer cells exhibit higher iron requirements than noncancer cells, treatment with ferroptosis-inducing compounds may be a feasible strategy for cancer therapy. However, cancer cells develop acquired resistance to evade ferroptosis, and the mechanisms responsible for ferroptosis resistance are not fully clarified. In the current study, we reported that DDX39B was downregulated during sorafenib-induced ferroptosis in a dose- and time-dependent manner. Exogenous introduction of DDX39B ensured the survival of HCC cells upon exposure to sorafenib, while the opposite phenomenon was observed in DDX39B-silenced HCC cells. Mechanistically, we demonstrated that DDX39B increased GPX4 levels by promoting the splicing and cytoplasmic translocation of GPX4 pre-mRNA, which was sufficient to detoxify sorafenib-triggered excess lipid ROS production, lipid peroxidation accumulation, ferrous iron levels, and mitochondrial damage. Inhibition of DDX39B ATPase activity by CCT018159 repressed the splicing and cytoplasmic export of GPX4 pre-mRNA and synergistically assisted sorafenib-induced ferroptotic cell death in HCC cells. Taken together, our data uncover a novel role for DDX39B in ferroptosis resistance by modulating the maturation of GPX4 mRNA via a posttranscriptional approach and suggest that DDX39B inhibition may be a promising therapeutic strategy to enhance the sensitivity and vulnerability of HCC cells to sorafenib.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , DEAD-box RNA Helicases , Ferroptosis , Liver Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , RNA Precursors , Sorafenib , Ferroptosis/drug effects , Ferroptosis/physiology , Sorafenib/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , Antineoplastic Agents/pharmacology , Animals , Mice , RNA Splicing/drug effects , Mice, Nude , Cell Line, Tumor , Dose-Response Relationship, Drug , Mice, Inbred BALB C , Male , Cytoplasm/metabolism , Cytoplasm/drug effects
7.
Nature ; 629(8014): 1165-1173, 2024 May.
Article in English | MEDLINE | ID: mdl-38720076

ABSTRACT

The nucleus is highly organized, such that factors involved in the transcription and processing of distinct classes of RNA are confined within specific nuclear bodies1,2. One example is the nuclear speckle, which is defined by high concentrations of protein and noncoding RNA regulators of pre-mRNA splicing3. What functional role, if any, speckles might play in the process of mRNA splicing is unclear4,5. Here we show that genes localized near nuclear speckles display higher spliceosome concentrations, increased spliceosome binding to their pre-mRNAs and higher co-transcriptional splicing levels than genes that are located farther from nuclear speckles. Gene organization around nuclear speckles is dynamic between cell types, and changes in speckle proximity lead to differences in splicing efficiency. Finally, directed recruitment of a pre-mRNA to nuclear speckles is sufficient to increase mRNA splicing levels. Together, our results integrate the long-standing observations of nuclear speckles with the biochemistry of mRNA splicing and demonstrate a crucial role for dynamic three-dimensional spatial organization of genomic DNA in driving spliceosome concentrations and controlling the efficiency of mRNA splicing.


Subject(s)
Genome , Nuclear Speckles , RNA Precursors , RNA Splicing , RNA, Messenger , Spliceosomes , Animals , Humans , Male , Mice , Genes , Genome/genetics , Human Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Nuclear Speckles/genetics , Nuclear Speckles/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spliceosomes/metabolism , Transcription, Genetic
8.
Nat Commun ; 15(1): 3839, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714659

ABSTRACT

Pre-mRNA splicing, a key process in gene expression, can be therapeutically modulated using various drug modalities, including antisense oligonucleotides (ASOs). However, determining promising targets is hampered by the challenge of systematically mapping splicing-regulatory elements (SREs) in their native sequence context. Here, we use the catalytically inactive CRISPR-RfxCas13d RNA-targeting system (dCas13d/gRNA) as a programmable platform to bind SREs and modulate splicing by competing against endogenous splicing factors. SpliceRUSH, a high-throughput screening method, was developed to map SREs in any gene of interest using a lentivirus gRNA library that tiles the genetic region, including distal intronic sequences. When applied to SMN2, a therapeutic target for spinal muscular atrophy, SpliceRUSH robustly identifies not only known SREs but also a previously unknown distal intronic SRE, which can be targeted to alter exon 7 splicing using either dCas13d/gRNA or ASOs. This technology enables a deeper understanding of splicing regulation with applications for RNA-based drug discovery.


Subject(s)
CRISPR-Cas Systems , Exons , Introns , RNA Splicing , RNA, Guide, CRISPR-Cas Systems , Survival of Motor Neuron 2 Protein , Humans , RNA Splicing/genetics , Survival of Motor Neuron 2 Protein/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , Introns/genetics , Exons/genetics , HEK293 Cells , Oligonucleotides, Antisense/genetics , Muscular Atrophy, Spinal/genetics , Regulatory Sequences, Nucleic Acid/genetics , RNA Precursors/genetics , RNA Precursors/metabolism
9.
Nat Commun ; 15(1): 4110, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750024

ABSTRACT

Maturation of eukaryotic pre-mRNAs via splicing and polyadenylation is modulated across cell types and conditions by a variety of RNA-binding proteins (RBPs). Although there exist over 1,500 RBPs in human cells, their binding motifs and functions still remain to be elucidated, especially in the complex environment of tissues and in the context of diseases. To overcome the lack of methods for the systematic and automated detection of sequence motif-guided pre-mRNA processing regulation from RNA sequencing (RNA-Seq) data we have developed MAPP (Motif Activity on Pre-mRNA Processing). Applying MAPP to RBP knock-down experiments reveals that many RBPs regulate both splicing and polyadenylation of nascent transcripts by acting on similar sequence motifs. MAPP not only infers these sequence motifs, but also unravels the position-dependent impact of the RBPs on pre-mRNA processing. Interestingly, all investigated RBPs that act on both splicing and 3' end processing exhibit a consistently repressive or activating effect on both processes, providing a first glimpse on the underlying mechanism. Applying MAPP to normal and malignant brain tissue samples unveils that the motifs bound by the PTBP1 and RBFOX RBPs coordinately drive the oncogenic splicing program active in glioblastomas demonstrating that MAPP paves the way for characterizing pre-mRNA processing regulators under physiological and pathological conditions.


Subject(s)
Polyadenylation , RNA Precursors , RNA Splicing , RNA-Binding Proteins , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Nucleotide Motifs , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics
10.
Nat Cell Biol ; 26(6): 946-961, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38745030

ABSTRACT

RNA-binding proteins (RBPs) are pivotal in acute myeloid leukaemia (AML), a lethal disease. Although specific phase separation-competent RBPs are recognized in AML, the effect of their condensate formation on AML leukaemogenesis, and the therapeutic potential of inhibition of phase separation are underexplored. In our in vivo CRISPR RBP screen, fibrillarin (FBL) emerges as a crucial nucleolar protein that regulates AML cell survival, primarily through its phase separation domains rather than methyltransferase or acetylation domains. These phase separation domains, with specific features, coordinately drive nucleoli formation and early processing of pre-rRNA (including efflux, cleavage and methylation), eventually enhancing the translation of oncogenes such as MYC. Targeting the phase separation capability of FBL with CGX-635 leads to elimination of AML cells, suggesting an additional mechanism of action for CGX-635 that complements its established therapeutic effects. We highlight the potential of PS modulation of critical proteins as a possible therapeutic strategy for AML.


Subject(s)
Chromosomal Proteins, Non-Histone , Leukemia, Myeloid, Acute , RNA Precursors , RNA Processing, Post-Transcriptional , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Animals , Cell Line, Tumor , Protein Biosynthesis , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Phase Separation
11.
Cancer Lett ; 594: 216980, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38797229

ABSTRACT

Acute myeloid leukemia (AML) is frequently linked to genetic abnormalities, with the t (8; 21) translocation, resulting in the production of a fusion oncoprotein AML1-ETO (AE), being a prevalent occurrence. This protein plays a pivotal role in t (8; 21) AML's onset, advancement, and recurrence, making it a therapeutic target. However, the development of drug molecules targeting AML1-ETO are markedly insufficient, especially used in clinical treatment. In this study, it was uncovered that Neratinib could significantly downregulate AML1-ETO protein level, subsequently promoting differentiation of t (8; 21) AML cells. Based on "differentiated active" probes, Neratinib was identified as a functional inhibitor against HNRNPA3 through covalent binding. The further studies demonstrated that HNRNPA3 function as a putative m6A reader responsible for recognizing and regulating the alternative splicing of AML-ETO pre-mRNA. These findings not only contribute to a novel insight to the mechanism governing post-transcriptional modification of AML1-ETO transcript, but also suggest that Neratinib would be promising therapeutic potential for t (8; 21) AML treatment.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , Oncogene Proteins, Fusion , Quinolines , RUNX1 Translocation Partner 1 Protein , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Quinolines/pharmacology , Cell Differentiation/drug effects , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Translocation, Genetic/drug effects , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Alternative Splicing/drug effects , Cell Line, Tumor , Animals , Mice
12.
Microbiol Spectr ; 12(7): e0351323, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38785434

ABSTRACT

Sweet orange (Citrus sinensis) is one of the most important fruit crops worldwide. Virus infections in this crop can interfere with cellular processes, causing dramatic economic losses. By performing RT-qPCR analyses, we demonstrated that citrus psorosis virus (CPsV)-infected orange plants exhibited higher levels of unprocessed microRNA (miRNA) precursors than healthy plants. This result correlated with the reported reduction of mature miRNAs species. The protein 24K, the CPsV suppressor of RNA silencing (VSR), interacts with miRNA precursors in vivo. Thus, this protein becomes a candidate responsible for the increased accumulation of unprocessed miRNAs. We analyzed 24K RNA-binding and protein-protein interaction domains and described patterns of its subcellular localization. We also showed that 24K colocalizes within nuclear D-bodies with the miRNA biogenesis proteins DICER-LIKE 1 (DCL1), HYPONASTIC LEAVES 1 (HYL1), and SERRATE (SE). According to the results of bimolecular fluorescence complementation and co-immunoprecipitation assays, the 24K protein interacts with HYL1 and SE. Thus, 24K may inhibit miRNA processing in CPsV-infected citrus plants by direct interaction with the miRNA processing complex. This work contributes to the understanding of how a virus can alter the regulatory mechanisms of the host, particularly miRNA biogenesis and function.IMPORTANCESweet oranges can suffer from disease symptoms induced by virus infections, thus resulting in drastic economic losses. In sweet orange plants, CPsV alters the accumulation of some precursors from the regulatory molecules called miRNAs. This alteration leads to a decreased level of mature miRNA species. This misregulation may be due to a direct association of one of the viral proteins (24K) with miRNA precursors. On the other hand, 24K may act with components of the cell miRNA processing machinery through a series of predicted RNA-binding and protein-protein interaction domains.


Subject(s)
Citrus sinensis , MicroRNAs , Plant Diseases , Viral Proteins , MicroRNAs/metabolism , MicroRNAs/genetics , Plant Diseases/virology , Viral Proteins/metabolism , Viral Proteins/genetics , Citrus sinensis/virology , Citrus sinensis/metabolism , Plant Viruses/genetics , Plant Viruses/metabolism , Plant Viruses/physiology , Plant Proteins/metabolism , Plant Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA Processing, Post-Transcriptional , Citrus/virology , Citrus/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics
13.
Proc Natl Acad Sci U S A ; 121(21): e2322974121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743621

ABSTRACT

SRSF1 is the founding member of the SR protein family. It is required-interchangeably with other SR proteins-for pre-mRNA splicing in vitro, and it regulates various alternative splicing events. Dysregulation of SRSF1 expression contributes to cancer and other pathologies. Here, we characterized SRSF1's interactome using proximity labeling and mass spectrometry. This approach yielded 190 proteins enriched in the SRSF1 samples, independently of the N- or C-terminal location of the biotin-labeling domain. The detected proteins reflect established functions of SRSF1 in pre-mRNA splicing and reveal additional connections to spliceosome proteins, in addition to other recently identified functions. We validated a robust interaction with the spliceosomal RNA helicase DDX23/PRP28 using bimolecular fluorescence complementation and in vitro binding assays. The interaction is mediated by the N-terminal RS-like domain of DDX23 and both RRM1 and the RS domain of SRSF1. During pre-mRNA splicing, DDX23's ATPase activity is essential for the pre-B to B spliceosome complex transition and for release of U1 snRNP from the 5' splice site. We show that the RS-like region of DDX23's N-terminal domain is important for spliceosome incorporation, while larger deletions in this domain alter subnuclear localization. We discuss how the identified interaction of DDX23 with SRSF1 and other SR proteins may be involved in the regulation of these processes.


Subject(s)
DEAD-box RNA Helicases , RNA Splicing , Serine-Arginine Splicing Factors , Spliceosomes , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Humans , Spliceosomes/metabolism , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , Protein Binding , HeLa Cells
14.
Nat Commun ; 15(1): 4622, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816438

ABSTRACT

The 5'-end capping of nascent pre-mRNA represents the initial step in RNA processing, with evidence demonstrating that guanosine addition and 2'-O-ribose methylation occur in tandem with early steps of transcription by RNA polymerase II, especially at the pausing stage. Here, we determine the cryo-EM structures of the paused elongation complex in complex with RNGTT, as well as the paused elongation complex in complex with RNGTT and CMTR1. Our findings show the simultaneous presence of RNGTT and the NELF complex bound to RNA polymerase II. The NELF complex exhibits two conformations, one of which shows a notable rearrangement of NELF-A/D compared to that of the paused elongation complex. Moreover, CMTR1 aligns adjacent to RNGTT on the RNA polymerase II stalk. Our structures indicate that RNGTT and CMTR1 directly bind the paused elongation complex, illuminating the mechanism by which 5'-end capping of pre-mRNA during transcriptional pausing.


Subject(s)
Cryoelectron Microscopy , RNA Caps , RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/chemistry , RNA Caps/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics , Humans , Protein Binding , Models, Molecular , RNA, Messenger/metabolism , RNA, Messenger/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry
15.
Cells ; 13(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38786046

ABSTRACT

Our study focused on assessing the effects of three newly identified BRCA1 exon 11 variants (c.1019T>C, c.2363T>G, and c.3192T>C) on breast cancer susceptibility. Using computational predictions and experimental splicing assays, we evaluated their potential as pathogenic mutations. Our in silico analyses suggested that the c.2363T>G and c.3192T>C variants could impact both splicing and protein function, resulting in the V340A and V788G mutations, respectively. We further examined their splicing effects using minigene assays in MCF7 and SKBR3 breast cancer cell lines. Interestingly, we found that the c.2363T>G variant significantly altered splicing patterns in MCF7 cells but not in SKBR3 cells. This finding suggests a potential influence of cellular context on the variant's effects. While attempts to correlate in silico predictions with RNA binding factors were inconclusive, this observation underscores the complexity of splicing regulation. Splicing is governed by various factors, including cellular contexts and protein interactions, making it challenging to predict outcomes accurately. Further research is needed to fully understand the functional consequences of the c.2363T>G variant in breast cancer pathogenesis. Integrating computational predictions with experimental data will provide valuable insights into the role of alternative splicing regulation in different breast cancer types and stages.


Subject(s)
BRCA1 Protein , Breast Neoplasms , Exons , RNA Precursors , RNA Splicing , Humans , Exons/genetics , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Cell Line, Tumor , Mutation/genetics , MCF-7 Cells , Alternative Splicing/genetics , Genetic Predisposition to Disease
16.
Sci Adv ; 10(19): eadn1547, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38718117

ABSTRACT

Pre-mRNA splicing is a fundamental step in gene expression, conserved across eukaryotes, in which the spliceosome recognizes motifs at the 3' and 5' splice sites (SSs), excises introns, and ligates exons. SS recognition and pairing is often influenced by protein splicing factors (SFs) that bind to splicing regulatory elements (SREs). Here, we describe SMsplice, a fully interpretable model of pre-mRNA splicing that combines models of core SS motifs, SREs, and exonic and intronic length preferences. We learn models that predict SS locations with 83 to 86% accuracy in fish, insects, and plants and about 70% in mammals. Learned SRE motifs include both known SF binding motifs and unfamiliar motifs, and both motif classes are supported by genetic analyses. Our comparisons across species highlight similarities between non-mammals, increased reliance on intronic SREs in plant splicing, and a greater reliance on SREs in mammalian splicing.


Subject(s)
Exons , Introns , RNA Precursors , RNA Splice Sites , RNA Splicing , RNA Precursors/genetics , RNA Precursors/metabolism , Animals , Introns/genetics , Exons/genetics , Genes, Plant , Models, Genetic , Spliceosomes/metabolism , Spliceosomes/genetics , Plants/genetics , Humans , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
17.
Nucleic Acids Res ; 52(11): 6360-6375, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38682589

ABSTRACT

Although DNA-PK inhibitors (DNA-PK-i) have been applied in clinical trials for cancer treatment, the biomarkers and mechanism of action of DNA-PK-i in tumor cell suppression remain unclear. Here, we observed that a low dose of DNA-PK-i and PARP inhibitor (PARP-i) synthetically suppresses BRCA-deficient tumor cells without inducing DNA double-strand breaks (DSBs). Instead, we found that a fraction of DNA-PK localized inside of nucleoli, where we did not observe obvious DSBs. Moreover, the Ku proteins recognize pre-rRNA that facilitates DNA-PKcs autophosphorylation independent of DNA damage. Ribosomal proteins are also phosphorylated by DNA-PK, which regulates pre-rRNA biogenesis. In addition, DNA-PK-i acts together with PARP-i to suppress pre-rRNA biogenesis and tumor cell growth. Collectively, our studies reveal a DNA damage repair-independent role of DNA-PK-i in tumor suppression.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase , Ku Autoantigen , RNA Precursors , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Humans , RNA Precursors/metabolism , RNA Precursors/genetics , Cell Line, Tumor , Ku Autoantigen/metabolism , Ku Autoantigen/genetics , Phosphorylation , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , Cell Nucleolus/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal/genetics , Animals , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
18.
Mol Syst Biol ; 20(6): 676-701, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38664594

ABSTRACT

Splice-switching oligonucleotides (SSOs) are antisense compounds that act directly on pre-mRNA to modulate alternative splicing (AS). This study demonstrates the value that artificial intelligence/machine learning (AI/ML) provides for the identification of functional, verifiable, and therapeutic SSOs. We trained XGboost tree models using splicing factor (SF) pre-mRNA binding profiles and spliceosome assembly information to identify modulatory SSO binding sites on pre-mRNA. Using Shapley and out-of-bag analyses we also predicted the identity of specific SFs whose binding to pre-mRNA is blocked by SSOs. This step adds considerable transparency to AI/ML-driven drug discovery and informs biological insights useful in further validation steps. We applied this approach to previously established functional SSOs to retrospectively identify the SFs likely to regulate those events. We then took a prospective validation approach using a novel target in triple negative breast cancer (TNBC), NEDD4L exon 13 (NEDD4Le13). Targeting NEDD4Le13 with an AI/ML-designed SSO decreased the proliferative and migratory behavior of TNBC cells via downregulation of the TGFß pathway. Overall, this study illustrates the ability of AI/ML to extract actionable insights from RNA-seq data.


Subject(s)
Alternative Splicing , Artificial Intelligence , Machine Learning , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Oligonucleotides, Antisense/genetics , Cell Movement/genetics , Spliceosomes/metabolism , Spliceosomes/genetics , Oligonucleotides/genetics , Female
19.
Nucleic Acids Res ; 52(8): 4483-4501, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587191

ABSTRACT

Messenger RNA precursors (pre-mRNA) generally undergo 3' end processing by cleavage and polyadenylation (CPA), which is specified by a polyadenylation site (PAS) and adjacent RNA sequences and regulated by a large variety of core and auxiliary CPA factors. To date, most of the human CPA factors have been discovered through biochemical and proteomic studies. However, genetic identification of the human CPA factors has been hampered by the lack of a reliable genome-wide screening method. We describe here a dual fluorescence readthrough reporter system with a PAS inserted between two fluorescent reporters. This system enables measurement of the efficiency of 3' end processing in living cells. Using this system in combination with a human genome-wide CRISPR/Cas9 library, we conducted a screen for CPA factors. The screens identified most components of the known core CPA complexes and other known CPA factors. The screens also identified CCNK/CDK12 as a potential core CPA factor, and RPRD1B as a CPA factor that binds RNA and regulates the release of RNA polymerase II at the 3' ends of genes. Thus, this dual fluorescence reporter coupled with CRISPR/Cas9 screens reliably identifies bona fide CPA factors and provides a platform for investigating the requirements for CPA in various contexts.


Subject(s)
CRISPR-Cas Systems , Genes, Reporter , RNA Precursors , mRNA Cleavage and Polyadenylation Factors , Humans , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Genome, Human , HEK293 Cells , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , Polyadenylation , RNA Cleavage , RNA Polymerase II/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics
20.
Int J Biol Macromol ; 268(Pt 1): 131781, 2024 May.
Article in English | MEDLINE | ID: mdl-38657924

ABSTRACT

Alternative splicing is a crucial regulator in stem cell biology, intricately influencing the functions of various biological macromolecules, particularly pre-mRNAs and the resultant protein isoforms. This regulatory mechanism is vital in determining stem cell pluripotency, differentiation, and proliferation. Alternative splicing's role in allowing single genes to produce multiple protein isoforms facilitates the proteomic diversity that is essential for stem cells' functional complexity. This review delves into the critical impact of alternative splicing on cellular functions, focusing on its interaction with key macromolecules and how this affects cellular behavior. We critically examine how alternative splicing modulates the function and stability of pre-mRNAs, leading to diverse protein expressions that govern stem cell characteristics, including pluripotency, self-renewal, survival, proliferation, differentiation, aging, migration, somatic reprogramming, and genomic stability. Furthermore, the review discusses the therapeutic potential of targeting alternative splicing-related pathways in disease treatment, particularly focusing on the modulation of RNA and protein interactions. We address the challenges and future prospects in this field, underscoring the need for further exploration to unravel the complex interplay between alternative splicing, RNA, proteins, and stem cell behaviors, which is crucial for advancing our understanding and therapeutic approaches in regenerative medicine and disease treatment.


Subject(s)
Alternative Splicing , RNA Precursors , Stem Cells , Humans , RNA Precursors/genetics , RNA Precursors/metabolism , Animals , Stem Cells/metabolism , Stem Cells/cytology , Cell Differentiation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...