Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 666
Filter
1.
Science ; 383(6688): 1245-1252, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38484052

ABSTRACT

The minor spliceosome, which is responsible for the splicing of U12-type introns, comprises five small nuclear RNAs (snRNAs), of which only one is shared with the major spliceosome. In this work, we report the 3.3-angstrom cryo-electron microscopy structure of the fully assembled human minor spliceosome pre-B complex. The atomic model includes U11 small nuclear ribonucleoprotein (snRNP), U12 snRNP, and U4atac/U6atac.U5 tri-snRNP. U11 snRNA is recognized by five U11-specific proteins (20K, 25K, 35K, 48K, and 59K) and the heptameric Sm ring. The 3' half of the 5'-splice site forms a duplex with U11 snRNA; the 5' half is recognized by U11-35K, U11-48K, and U11 snRNA. Two proteins, CENATAC and DIM2/TXNL4B, specifically associate with the minor tri-snRNP. A structural analysis uncovered how two conformationally similar tri-snRNPs are differentiated by the minor and major prespliceosomes for assembly.


Subject(s)
Introns , RNA, Small Nuclear , Spliceosomes , Humans , Cryoelectron Microscopy , Ribonucleoproteins, Small Nuclear/chemistry , RNA Splice Sites , RNA Splicing , RNA, Small Nuclear/chemistry , Spliceosomes/chemistry , Nucleic Acid Conformation
2.
Nucleic Acids Res ; 52(7): 4037-4052, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38499487

ABSTRACT

Here, we identify RBM41 as a novel unique protein component of the minor spliceosome. RBM41 has no previously recognized cellular function but has been identified as a paralog of U11/U12-65K, a known unique component of the U11/U12 di-snRNP. Both proteins use their highly similar C-terminal RRMs to bind to 3'-terminal stem-loops in U12 and U6atac snRNAs with comparable affinity. Our BioID data indicate that the unique N-terminal domain of RBM41 is necessary for its association with complexes containing DHX8, an RNA helicase, which in the major spliceosome drives the release of mature mRNA from the spliceosome. Consistently, we show that RBM41 associates with excised U12-type intron lariats, is present in the U12 mono-snRNP, and is enriched in Cajal bodies, together suggesting that RBM41 functions in the post-splicing steps of the minor spliceosome assembly/disassembly cycle. This contrasts with U11/U12-65K, which uses its N-terminal region to interact with U11 snRNP during intron recognition. Finally, while RBM41 knockout cells are viable, they show alterations in U12-type 3' splice site usage. Together, our results highlight the role of the 3'-terminal stem-loop of U12 snRNA as a dynamic binding platform for the U11/U12-65K and RBM41 proteins, which function at distinct stages of the assembly/disassembly cycle.


Subject(s)
DEAD-box RNA Helicases , RNA Splicing Factors , RNA, Small Nuclear , RNA-Binding Proteins , Ribonucleoproteins, Small Nuclear , Spliceosomes , Spliceosomes/metabolism , Spliceosomes/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Humans , RNA, Small Nuclear/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/chemistry , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA Splicing , Introns/genetics , HeLa Cells , Protein Binding , Coiled Bodies/metabolism , HEK293 Cells
3.
Nat Struct Mol Biol ; 31(5): 835-845, 2024 May.
Article in English | MEDLINE | ID: mdl-38196034

ABSTRACT

Selection of the pre-mRNA branch site (BS) by the U2 small nuclear ribonucleoprotein (snRNP) is crucial to prespliceosome (A complex) assembly. The RNA helicase PRP5 proofreads BS selection but the underlying mechanism remains unclear. Here we report the atomic structures of two sequential complexes leading to prespliceosome assembly: human 17S U2 snRNP and a cross-exon pre-A complex. PRP5 is anchored on 17S U2 snRNP mainly through occupation of the RNA path of SF3B1 by an acidic loop of PRP5; the helicase domain of PRP5 associates with U2 snRNA; the BS-interacting stem-loop (BSL) of U2 snRNA is shielded by TAT-SF1, unable to engage the BS. In the pre-A complex, an initial U2-BS duplex is formed; the translocated helicase domain of PRP5 stays with U2 snRNA and the acidic loop still occupies the RNA path. The pre-A conformation is specifically stabilized by the splicing factors SF1, DNAJC8 and SF3A2. Cancer-derived mutations in SF3B1 damage its association with PRP5, compromising BS proofreading. Together, these findings reveal key insights into prespliceosome assembly and BS selection or proofreading by PRP5.


Subject(s)
Models, Molecular , RNA Splicing Factors , Spliceosomes , Humans , Spliceosomes/metabolism , Spliceosomes/chemistry , RNA Splicing Factors/metabolism , RNA Splicing Factors/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/genetics , Cryoelectron Microscopy , RNA Splicing , RNA Precursors/metabolism , Nucleic Acid Conformation , RNA, Small Nuclear/metabolism , RNA, Small Nuclear/chemistry , Phosphoproteins
4.
Commun Biol ; 5(1): 819, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35970937

ABSTRACT

Productive transcriptional elongation of many cellular and viral mRNAs requires transcriptional factors to extract pTEFb from the 7SK snRNP by modulating the association between HEXIM and 7SK snRNA. In HIV-1, Tat binds to 7SK by displacing HEXIM. However, without the structure of the 7SK-HEXIM complex, the constraints that must be overcome for displacement remain unknown. Furthermore, while structure details of the TatNL4-3-7SK complex have been elucidated, it is unclear how subtypes with more HEXIM-like Tat sequences accomplish displacement. Here we report the structures of HEXIM, TatG, and TatFin arginine rich motifs in complex with the apical stemloop-1 of 7SK. While most interactions between 7SK with HEXIM and Tat are similar, critical differences exist that guide function. First, the conformational plasticity of 7SK enables the formation of three different base pair configurations at a critical remodeling site, which allows for the modulation required for HEXIM binding and its subsequent displacement by Tat. Furthermore, the specific sequence variations observed in various Tat subtypes all converge on remodeling 7SK at this region. Second, we show that HEXIM primes its own displacement by causing specific local destabilization upon binding - a feature that is then exploited by Tat to bind 7SK more efficiently.


Subject(s)
HIV-1 , RNA-Binding Proteins , HIV-1/genetics , Nucleic Acid Conformation , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
5.
J Mol Biol ; 434(11): 167578, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35398145

ABSTRACT

Expression of mRNA is often regulated by the binding of a small RNA (miRNA, snoRNA, siRNA). While the pairing contribution to the net free energy is well parameterized and can be computed in O(N) time, the cost of removing pre-existing mRNA secondary structure has not received sufficient attention. Conventional methods for computing the unfolding free energy of a target mRNA are costly, scaling like the cube of the number of target bases O(N3). Here we introduce a model to describe the unfolding costs of the binding site, which features surprisingly big differences in the free energy parameters for the four bases. The model is implemented in our O(N) algorithm, BindOligoNet. Donor splice site prediction is more accurate when using our calculation of spliceosomal U1-snRNA to mRNA net binding free energy. Our base-dependent free energies also correlate with efficient ribosome docking near the start codon.


Subject(s)
Peptide Chain Initiation, Translational , RNA Splicing , RNA, Messenger , Algorithms , Binding Sites , Nucleic Acid Conformation , Nucleotides , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Small Nuclear/chemistry , Spliceosomes/chemistry , Thermodynamics
6.
Mol Cancer ; 21(1): 17, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35033060

ABSTRACT

BACKGROUND: miRNAs are regulatory transcripts established as repressors of mRNA stability and translation that have been functionally implicated in carcinogenesis. miR-10b is one of the key onco-miRs associated with multiple forms of cancer. Malignant gliomas exhibit particularly striking dependence on miR-10b. However, despite the therapeutic potential of miR-10b targeting, this miRNA's poorly investigated and largely unconventional properties hamper the clinical translation. METHODS: We utilized Covalent Ligation of Endogenous Argonaute-bound RNAs and their high-throughput RNA sequencing to identify miR-10b interactome and a combination of biochemical and imaging approaches for target validation. They included Crosslinking and RNA immunoprecipitation with spliceosomal proteins, a combination of miRNA FISH with protein immunofluorescence in glioma cells and patient-derived tumors, native Northern blotting, and the transcriptome-wide analysis of alternative splicing. RESULTS: We demonstrate that miR-10b binds to U6 snRNA, a core component of the spliceosomal machinery. We provide evidence of the direct binding between miR-10b and U6, in situ imaging of miR-10b and U6 co-localization in glioma cells and tumors, and biochemical co-isolation of miR-10b with the components of the spliceosome. We further demonstrate that miR-10b modulates U6 N-6-adenosine methylation and pseudouridylation, U6 binding to splicing factors SART3 and PRPF8, and regulates U6 stability, conformation, and levels. These effects on U6 result in global splicing alterations, exemplified by the altered ratio of the isoforms of a small GTPase CDC42, reduced overall CDC42 levels, and downstream CDC42 -mediated effects on cell viability. CONCLUSIONS: We identified U6 snRNA, the key RNA component of the spliceosome, as the top miR-10b target in glioblastoma. We, therefore, present an unexpected intersection of the miRNA and splicing machineries and a new nuclear function for a major cancer-associated miRNA.


Subject(s)
Cell Nucleus/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Oncogenes , RNA Splicing , RNA, Small Nuclear/genetics , Alternative Splicing , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Humans , Membrane Glycoproteins/genetics , Models, Biological , RNA Interference , RNA, Small Nuclear/chemistry , RNA-Binding Proteins/metabolism , Receptors, Immunologic/genetics , Spliceosomes/metabolism , cdc42 GTP-Binding Protein/genetics
7.
EMBO J ; 41(1): e107640, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34779515

ABSTRACT

SRSF1 protein and U1 snRNPs are closely connected splicing factors. They both stimulate exon inclusion, SRSF1 by binding to exonic splicing enhancer sequences (ESEs) and U1 snRNPs by binding to the downstream 5' splice site (SS), and both factors affect 5' SS selection. The binding of U1 snRNPs initiates spliceosome assembly, but SR proteins such as SRSF1 can in some cases substitute for it. The mechanistic basis of this relationship is poorly understood. We show here by single-molecule methods that a single molecule of SRSF1 can be recruited by a U1 snRNP. This reaction is independent of exon sequences and separate from the U1-independent process of binding to an ESE. Structural analysis and cross-linking data show that SRSF1 contacts U1 snRNA stem-loop 3, which is required for splicing. We suggest that the recruitment of SRSF1 to a U1 snRNP at a 5'SS is the basis for exon definition by U1 snRNP and might be one of the principal functions of U1 snRNPs in the core reactions of splicing in mammals.


Subject(s)
Exons/genetics , Nucleic Acid Conformation , Ribonucleoprotein, U1 Small Nuclear/metabolism , Serine-Arginine Splicing Factors/metabolism , HeLa Cells , Humans , Models, Biological , Protein Binding , RNA Precursors/metabolism , RNA Splice Sites/genetics , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism
8.
Science ; 375(6576): 50-57, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34822310

ABSTRACT

Recognition of the intron branch site (BS) by the U2 small nuclear ribonucleoprotein (snRNP) is a critical event during spliceosome assembly. In mammals, BS sequences are poorly conserved, and unambiguous intron recognition cannot be achieved solely through a base-pairing mechanism. We isolated human 17S U2 snRNP and reconstituted in vitro its adenosine 5´-triphosphate (ATP)­dependent remodeling and binding to the pre­messenger RNA substrate. We determined a series of high-resolution (2.0 to 2.2 angstrom) structures providing snapshots of the BS selection process. The substrate-bound U2 snRNP shows that SF3B6 stabilizes the BS:U2 snRNA duplex, which could aid binding of introns with poor sequence complementarity. ATP-dependent remodeling uncoupled from substrate binding captures U2 snRNA in a conformation that competes with BS recognition, providing a selection mechanism based on branch helix stability.


Subject(s)
Introns , RNA Precursors/chemistry , Ribonucleoprotein, U2 Small Nuclear/chemistry , Spliceosomes/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , Nucleic Acid Conformation , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Conformation , RNA Precursors/metabolism , RNA Splicing , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spliceosomes/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism
9.
PLoS One ; 16(10): e0258551, 2021.
Article in English | MEDLINE | ID: mdl-34648557

ABSTRACT

U2 snRNP is an essential component of the spliceosome. It is responsible for branch point recognition in the spliceosome A-complex via base-pairing of U2 snRNA with an intron to form the branch helix. Small molecule inhibitors target the SF3B component of the U2 snRNP and interfere with A-complex formation during spliceosome assembly. We previously found that the first SF3B inhibited-complex is less stable than A-complex and hypothesized that SF3B inhibitors interfere with U2 snRNA secondary structure changes required to form the branch helix. Using RNA chemical modifiers, we probed U2 snRNA structure in A-complex and SF3B inhibited splicing complexes. The reactivity pattern for U2 snRNA in the SF3B inhibited-complex is indistinguishable from that of A-complex, suggesting that they have the same secondary structure conformation, including the branch helix. This observation suggests SF3B inhibited-complex instability does not stem from an alternate RNA conformation and instead points to the inhibitors interfering with protein component interactions that normally stabilize U2 snRNP's association with an intron. In addition, we probed U2 snRNA in the free U2 snRNP in the presence of SF3B inhibitor and again saw no differences. However, increased protection of nucleotides upstream of Stem I in the absence of SF3A and SF3B proteins suggests a change of secondary structure at the very 5' end of U2 snRNA. Chemical probing of synthetic U2 snRNA in the absence of proteins results in similar protections and predicts a previously uncharacterized extension of Stem I. Because this stem must be disrupted for SF3A and SF3B proteins to stably join the snRNP, the structure has the potential to influence snRNP assembly and recycling after spliceosome disassembly.


Subject(s)
RNA, Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism , HeLa Cells , Humans , Nucleic Acid Conformation , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Ribonucleoprotein, U2 Small Nuclear/chemistry , Spliceosomes/metabolism
10.
J Mol Biol ; 433(21): 167229, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34487791

ABSTRACT

Although RNA-binding proteins (RBPs) are known to be enriched in intrinsic disorder, no previous analysis focused on RBPs interacting with specific RNA types. We fill this gap with a comprehensive analysis of the putative disorder in RBPs binding to six common RNA types: messenger RNA (mRNA), transfer RNA (tRNA), small nuclear RNA (snRNA), non-coding RNA (ncRNA), ribosomal RNA (rRNA), and internal ribosome RNA (irRNA). We also analyze the amount of putative intrinsic disorder in the RNA-binding domains (RBDs) and non-RNA-binding-domain regions (non-RBD regions). Consistent with previous studies, we show that in comparison with human proteome, RBPs are significantly enriched in disorder. However, closer examination finds significant enrichment in predicted disorder for the mRNA-, rRNA- and snRNA-binding proteins, while the proteins that interact with ncRNA and irRNA are not enriched in disorder, and the tRNA-binding proteins are significantly depleted in disorder. We show a consistent pattern of significant disorder enrichment in the non-RBD regions coupled with low levels of disorder in RBDs, which suggests that disorder is relatively rarely utilized in the RNA-binding regions. Our analysis of the non-RBD regions suggests that disorder harbors posttranslational modification sites and is involved in the putative interactions with DNA. Importantly, we utilize experimental data from DisProt and independent data from Pfam to validate the above observations that rely on the disorder predictions. This study provides new insights into the distribution of disorder across proteins that bind different RNA types and the functional role of disorder in the regions where it is enriched.


Subject(s)
Intrinsically Disordered Proteins/chemistry , RNA, Messenger/chemistry , RNA, Ribosomal/chemistry , RNA, Small Nuclear/chemistry , RNA, Transfer/chemistry , RNA, Untranslated/chemistry , RNA-Binding Proteins/chemistry , Acetylation , Binding Sites , Gene Expression , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Methylation , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Proteome/genetics , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ubiquitination
11.
RNA ; 27(10): 1186-1203, 2021 10.
Article in English | MEDLINE | ID: mdl-34234030

ABSTRACT

Human pre-mRNA splicing is primarily catalyzed by the major spliceosome, comprising five small nuclear ribonucleoprotein complexes, U1, U2, U4, U5, and U6 snRNPs, each of which contains the corresponding U-rich snRNA. These snRNAs are encoded by large gene families exhibiting significant sequence variation, but it remains unknown if most human snRNA genes are untranscribed pseudogenes or produce variant snRNAs with the potential to differentially influence splicing. Since gene duplication and variation are powerful mechanisms of evolutionary adaptation, we sought to address this knowledge gap by systematically profiling human U1, U2, U4, and U5 snRNA variant gene transcripts. We identified 55 transcripts that are detectably expressed in human cells, 38 of which incorporate into snRNPs and spliceosomes in 293T cells. All U1 snRNA variants are more than 1000-fold less abundant in spliceosomes than the canonical U1, whereas at least 1% of spliceosomes contain a variant of U2 or U4. In contrast, eight U5 snRNA sequence variants occupy spliceosomes at levels of 1% to 46%. Furthermore, snRNA variants display distinct expression patterns across five human cell lines and adult and fetal tissues. Different RNA degradation rates contribute to the diverse steady state levels of snRNA variants. Our findings suggest that variant spliceosomes containing noncanonical snRNAs may contribute to different tissue- and cell-type-specific alternative splicing patterns.


Subject(s)
RNA Splicing , RNA, Messenger/genetics , RNA, Small Nuclear/genetics , Spliceosomes/genetics , Adult , Base Pairing , Base Sequence , Cell Fractionation/methods , Exons , Fetus , HEK293 Cells , Humans , Introns , Molecular Sequence Annotation , Nucleic Acid Conformation , Organ Specificity , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Spliceosomes/chemistry , Spliceosomes/metabolism
12.
ChemMedChem ; 16(20): 3181-3184, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34233081

ABSTRACT

The 332-nucleotide small nuclear RNA (snRNA) 7SK is a highly conserved non-coding RNA that regulates transcriptional elongation. By binding with positive transcriptional elongation factor b (P-TEFb) via HEXIM1, 7SK snRNA decreases the kinase activity of P-TEFb and inhibits transcriptional elongation. Additionally, it is reported that 7SK inhibition results in the stimulation of human immunodeficiency virus (HIV)-specific transcription. These reports suggest that 7SK is a naturally occurring functional molecule as negative regulator of P-TEFb and HIV transcription. In this study, we developed functional oligonucleotides that mimic the function of 7SK (7SK mimics) as novel inhibitors of HIV replication. We defined the essential region of 7SK regarding its suppressive effects on transcriptional downregulation using an antisense strategy. Based on the results, we designed 7SK mimics containing the defined region. The inhibitory effects of 7SK mimics on HIV-1 long terminal repeat promoter specific transcription was drastic compared with those of the control mimic molecule. Notably, these effects were found to be more enhanced by co-transfection with Tat-expressing plasmids. From these results, it is indicated that 7SK mimics may have great therapeutic potential for HIV/AIDS treatment.


Subject(s)
Drug Development , RNA, Small Nuclear/pharmacology , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Dose-Response Relationship, Drug , Molecular Structure , RNA, Small Nuclear/chemical synthesis , RNA, Small Nuclear/chemistry , Structure-Activity Relationship , Transcription, Genetic/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics
13.
Genes Dev ; 35(15-16): 1123-1141, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34301768

ABSTRACT

Spliceosomal small nuclear RNAs (snRNAs) are modified by small Cajal body (CB)-specific ribonucleoproteins (scaRNPs) to ensure snRNP biogenesis and pre-mRNA splicing. However, the function and subcellular site of snRNA modification are largely unknown. We show that CB localization of the protein Nopp140 is essential for concentration of scaRNPs in that nuclear condensate; and that phosphorylation by casein kinase 2 (CK2) at ∼80 serines targets Nopp140 to CBs. Transiting through CBs, snRNAs are apparently modified by scaRNPs. Indeed, Nopp140 knockdown-mediated release of scaRNPs from CBs severely compromises 2'-O-methylation of spliceosomal snRNAs, identifying CBs as the site of scaRNP catalysis. Additionally, alternative splicing patterns change indicating that these modifications in U1, U2, U5, and U12 snRNAs safeguard splicing fidelity. Given the importance of CK2 in this pathway, compromised splicing could underlie the mode of action of small molecule CK2 inhibitors currently considered for therapy in cholangiocarcinoma, hematological malignancies, and COVID-19.


Subject(s)
Interstitial Cells of Cajal/metabolism , Methylation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Splicing , RNA, Small Nuclear/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cholangiocarcinoma/drug therapy , Hematologic Neoplasms/drug therapy , Humans , Phosphorylation , RNA, Small Nuclear/chemistry , Ribonucleoproteins/metabolism , Spliceosomes/genetics , COVID-19 Drug Treatment
14.
RNA Biol ; 18(12): 2576-2593, 2021 12.
Article in English | MEDLINE | ID: mdl-34105434

ABSTRACT

During spliceosome assembly, interactions that bring the 5' and 3' ends of an intron in proximity are critical for the production of mature mRNA. Here, we report synergistic roles for the stem-loops 3 (SL3) and 4 (SL4) of the human U1 small nuclear RNA (snRNA) in maintaining the optimal U1 snRNP function, and formation of cross-intron contact with the U2 snRNP. We find that SL3 and SL4 bind distinct spliceosomal proteins and combining a U1 snRNA activity assay with siRNA-mediated knockdown, we demonstrate that SL3 and SL4 act through the RNA helicase UAP56 and the U2 protein SF3A1, respectively. In vitro analysis using UV crosslinking and splicing assays indicated that SL3 likely promotes the SL4-SF3A1 interaction leading to enhancement of A complex formation and pre-mRNA splicing. Overall, these results highlight the vital role of the distinct contacts of SL3 and SL4 in bridging the pre-mRNA bound U1 and U2 snRNPs during the early steps of human spliceosome assembly.


Subject(s)
Nucleic Acid Conformation , RNA Precursors/genetics , RNA Splicing , RNA, Messenger/genetics , RNA, Small Nuclear/genetics , Base Sequence , Humans , Introns , RNA Precursors/chemistry , RNA, Messenger/chemistry , RNA, Small Nuclear/chemistry
15.
Hum Mutat ; 42(8): 1042-1052, 2021 08.
Article in English | MEDLINE | ID: mdl-34085356

ABSTRACT

CDAGS Syndrome is a rare congenital disorder characterized by Craniosynostosis, Delayed closure of the fontanelles, cranial defects, clavicular hypoplasia, Anal and Genitourinary malformations, and Skin manifestations. We performed whole exome and Sanger sequencing to identify the underlying molecular cause in five patients with CDAGS syndrome from four distinct families. Whole exome sequencing revealed biallelic rare variants that disrupt highly conserved nucleotides within the RNU12 gene. RNU12 encodes a small nuclear RNA that is a component of the minor spliceosome and is essential for minor intron splicing. Targeted sequencing confirmed allele segregation within the four families. All five patients shared the same rare mutation NC_000022.10:g.43011402C>T, which alters a highly conserved nucleotide within the precursor U12 snRNA 3' extension. Each of them also carried a rare variant on the other allele that either disrupts the secondary structure or the Sm binding site of the RNU12 snRNA. Whole transcriptome sequencing analysis of lymphoblastoid cells identified 120 differentially expressed genes, and differential alternative splicing analysis indicated there was an enrichment of alternative splicing events in the patient. These findings provide evidence of the involvement of RNU12 in craniosynostosis, anal and genitourinary patterning, and cutaneous disease.


Subject(s)
Craniosynostoses , Digestive System Abnormalities , Porokeratosis , RNA, Small Nuclear/genetics , Anal Canal/abnormalities , Craniosynostoses/genetics , Humans , RNA Splicing , RNA, Small Nuclear/chemistry
16.
Science ; 372(6543)2021 05 14.
Article in English | MEDLINE | ID: mdl-33986153

ABSTRACT

Aberrant alternative splicing is a hallmark of cancer, yet the underlying regulatory programs that control this process remain largely unknown. Here, we report a systematic effort to decipher the RNA structural code that shapes pathological splicing during breast cancer metastasis. We discovered a previously unknown structural splicing enhancer that is enriched near cassette exons with increased inclusion in highly metastatic cells. We show that the spliceosomal protein small nuclear ribonucleoprotein polypeptide A' (SNRPA1) interacts with these enhancers to promote cassette exon inclusion. This interaction enhances metastatic lung colonization and cancer cell invasion, in part through SNRPA1-mediated regulation of PLEC alternative splicing, which can be counteracted by splicing modulating morpholinos. Our findings establish a noncanonical regulatory role for SNRPA1 as a prometastatic splicing enhancer in breast cancer.


Subject(s)
Alternative Splicing , Breast Neoplasms/pathology , Neoplasm Metastasis/genetics , RNA/genetics , RNA/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Adaptor Proteins, Signal Transducing/genetics , Algorithms , Animals , Binding Sites , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Progression , Exons , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Nucleic Acid Conformation , Plectin/genetics , Protein Binding , RNA Interference , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , RNA-Seq , Ribonucleoprotein, U2 Small Nuclear/genetics , Software , Spliceosomes/metabolism , Tumor Suppressor Proteins/genetics
17.
J Mol Biol ; 433(9): 166885, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33684393

ABSTRACT

7SK small nuclear RNA (snRNA) is an abundant and ubiquitously expressed noncoding RNA that functions to modulate the activity of RNA Polymerase II (RNAPII) in part by stabilizing distinct pools of 7SK-protein complexes. Prevailing models suggest that the secondary structure of 7SK is dynamically remodeled within its alternative RNA-protein pools such that its architecture differentially regulates the exchange of cognate binding partners. The nuclear hnRNP A1/A2 proteins influence the biology of 7SK snRNA via processes that require an intact stem loop (SL) 3 domain; however, the molecular details by which hnRNPs assemble onto 7SK snRNA are yet to be described. Here, we have taken an integrated approach to present a detailed description of the 7SK-hnRNP A1 complex. We show that unbound 7SK snRNA adopts at least two major conformations in solution, with significant structural differences localizing to the SL2-3 linker and the base of SL3. Phylogenetic analysis indicates that this same region is the least genetically conserved feature of 7SK snRNA. By performing DMS modifications with the presence of excess protein, we reveal that hnRNP A1 binds with selectivity to SL3 through mechanisms that increase the flexibility of the RNA adjacent to putative binding sites. Calorimetric titrations further validate that hnRNP A1-SL3 assembly is complex with the affinity of discrete binding events modulated by the surrounding RNA structure. To interpret this context-dependent binding phenomenon, we determined a 3D model of SL3 to show that it folds to position minimal hnRNP A1/A2 binding sites (5'-Y/RAG-3') within different local environments. SL3-protein complexes resolved by SEC-MALS-SAXS confirm that up to four hnRNP A1 proteins bind along the entire surface of SL3 via interactions that preserve the overall structural integrity of this domain. In sum, the collective results presented here reveal a specific role for a folded SL3 domain to scaffold hnRNP A1/A2-7SK assembly via mechanisms modulated by the surrounding RNA structure.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Nucleic Acid Conformation , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Animals , Base Sequence , Binding Sites , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding , Substrate Specificity
18.
Nucleic Acids Res ; 49(5): 2835-2847, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33577674

ABSTRACT

Disruption of minor spliceosome functions underlies several genetic diseases with mutations in the minor spliceosome-specific small nuclear RNAs (snRNAs) and proteins. Here, we define the molecular outcome of the U12 snRNA mutation (84C>U) resulting in an early-onset form of cerebellar ataxia. To understand the molecular consequences of the U12 snRNA mutation, we created cell lines harboring the 84C>T mutation in the U12 snRNA gene (RNU12). We show that the 84C>U mutation leads to accelerated decay of the snRNA, resulting in significantly reduced steady-state U12 snRNA levels. Additionally, the mutation leads to accumulation of 3'-truncated forms of U12 snRNA, which have undergone the cytoplasmic steps of snRNP biogenesis. Our data suggests that the 84C>U-mutant snRNA is targeted for decay following reimport into the nucleus, and that the U12 snRNA fragments are decay intermediates that result from the stalling of a 3'-to-5' exonuclease. Finally, we show that several other single-nucleotide variants in the 3' stem-loop of U12 snRNA that are segregating in the human population are also highly destabilizing. This suggests that the 3' stem-loop is important for the overall stability of the U12 snRNA and that additional disease-causing mutations are likely to exist in this region.


Subject(s)
Cerebellar Ataxia/genetics , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , HEK293 Cells , HeLa Cells , Humans , Mutation , Point Mutation , RNA Stability , RNA, Small Nuclear/metabolism
19.
Science ; 371(6535)2021 03 19.
Article in English | MEDLINE | ID: mdl-33509932

ABSTRACT

The minor spliceosome mediates splicing of the rare but essential U12-type precursor messenger RNA. Here, we report the atomic features of the activated human minor spliceosome determined by cryo-electron microscopy at 2.9-angstrom resolution. The 5' splice site and branch point sequence of the U12-type intron are recognized by the U6atac and U12 small nuclear RNAs (snRNAs), respectively. Five newly identified proteins stabilize the conformation of the catalytic center: The zinc finger protein SCNM1 functionally mimics the SF3a complex of the major spliceosome, the RBM48-ARMC7 complex binds the γ-monomethyl phosphate cap at the 5' end of U6atac snRNA, the U-box protein PPIL2 coordinates loop I of U5 snRNA and stabilizes U5 small nuclear ribonucleoprotein (snRNP), and CRIPT stabilizes U12 snRNP. Our study provides a framework for the mechanistic understanding of the function of the human minor spliceosome.


Subject(s)
Spliceosomes/chemistry , Spliceosomes/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/metabolism , Cryoelectron Microscopy , Cyclophilins/chemistry , Cyclophilins/metabolism , Humans , Introns , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , Protein Domains , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA Splicing , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA, Small Nuclear/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism
20.
RNA Biol ; 18(8): 1152-1159, 2021 08.
Article in English | MEDLINE | ID: mdl-33103602

ABSTRACT

Bacterial small-RNA (sRNA) sequences are functional RNAs, which play an important role in regulating the expression of a diverse class of genes. It is thus critical to identify such sRNA sequences and their probable mRNA targets. Here, we discuss new procedures to identify and characterize sRNA and their targets via the introduction of an integrated online platform 'PresRAT'. PresRAT uses the primary and secondary structural attributes of sRNA sequences to predict sRNA from a given sequence or bacterial genome. PresRAT also finds probable target mRNAs of sRNA sequences from a given bacterial chromosome and further concentrates on the identification of the probable sRNA-mRNA binding regions. Using PresRAT, we have identified a total of 66,209 potential sRNA sequences from 292 bacterial genomes and 2247 potential targets from 13 bacterial genomes. We have also implemented a protocol to build and refine 3D models of sRNA and sRNA-mRNA duplex regions and generated 3D models of 50 known sRNAs and 81 sRNA-mRNA duplexes using this platform. Along with the server part, PresRAT also contains a database section, which enlists the predicted sRNA sequences, sRNA targets, and their corresponding 3D models with structural dynamics information.


Subject(s)
Bacteria/genetics , RNA, Bacterial/chemistry , RNA, Messenger/chemistry , RNA, Small Cytoplasmic/chemistry , RNA, Small Nuclear/chemistry , Software , Bacteria/metabolism , Base Pairing , Benchmarking , Chromosomes, Bacterial/chemistry , Databases, Nucleic Acid , Nucleic Acid Conformation , Nucleic Acid Hybridization , RNA, Bacterial/classification , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/classification , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , RNA, Small Nuclear/classification , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...