Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Cell ; 172(3): 454-464.e11, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29361316

ABSTRACT

The spliceosome is a highly dynamic macromolecular complex that precisely excises introns from pre-mRNA. Here we report the cryo-EM 3D structure of the human Bact spliceosome at 3.4 Å resolution. In the Bact state, the spliceosome is activated but not catalytically primed, so that it is functionally blocked prior to the first catalytic step of splicing. The spliceosomal core is similar to the yeast Bact spliceosome; important differences include the presence of the RNA helicase aquarius and peptidyl prolyl isomerases. To examine the overall dynamic behavior of the purified spliceosome, we developed a principal component analysis-based approach. Calculating the energy landscape revealed eight major conformational states, which we refined to higher resolution. Conformational differences of the highly flexible structural components between these eight states reveal how spliceosomal components contribute to the assembly of the spliceosome, allowing it to generate a dynamic interaction network required for its subsequent catalytic activation.


Subject(s)
Molecular Dynamics Simulation , Spliceosomes/chemistry , HeLa Cells , Humans , Spliceosomes/metabolism , Spliceosomes/ultrastructure
2.
Cell ; 170(4): 701-713.e11, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28781166

ABSTRACT

Little is known about the spliceosome's structure before its extensive remodeling into a catalytically active complex. Here, we report a 3D cryo-EM structure of a pre-catalytic human spliceosomal B complex. The U2 snRNP-containing head domain is connected to the B complex main body via three main bridges. U4/U6.U5 tri-snRNP proteins, which are located in the main body, undergo significant rearrangements during tri-snRNP integration into the B complex. These include formation of a partially closed Prp8 conformation that creates, together with Dim1, a 5' splice site (ss) binding pocket, displacement of Sad1, and rearrangement of Brr2 such that it contacts its U4/U6 substrate and is poised for the subsequent spliceosome activation step. The molecular organization of several B-specific proteins suggests that they are involved in negatively regulating Brr2, positioning the U6/5'ss helix, and stabilizing the B complex structure. Our results indicate significant differences between the early activation phase of human and yeast spliceosomes.


Subject(s)
Spliceosomes/chemistry , Cell Nucleus/chemistry , Cryoelectron Microscopy , HeLa Cells , Humans , Models, Molecular , RNA-Binding Proteins/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Saccharomyces cerevisiae/chemistry , Spliceosomes/ultrastructure
3.
Cell ; 162(2): 456-456.e1, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186196

ABSTRACT

Numerous mechanisms exploit or modulate the conformational/compositional dynamics of spliceosomes to regulate splicing. The majority of higher eukaryotic protein-coding genes contain more than one intron and the derived pre-mRNAs can be alternatively spliced. Diverse principles ensure the reliable identification of authentic splice sites while concomitantly providing flexibility in splice site choice during alternative splicing. Some species contain a second type of minor (U12-type) spliceosome.


Subject(s)
RNA Splicing , Spliceosomes/metabolism , Animals , Humans , Ribonucleoproteins/metabolism , Spliceosomes/chemistry
4.
Cell ; 162(3): 690-690.e1, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232231

ABSTRACT

The complex compositional and conformational dynamics of spliceosomes required for regulated splicing are prone to malfunction when mutations affect splicing factors or cis-acting regulatory sequences. Indeed, many such mutations have been linked to heritable diseases or malignancies in humans. Small molecule modulators and antisense oligonucleotides or analogs harbor great potential for therapies and several substances that can modulate splicing events have entered clinical trials.


Subject(s)
Disease/genetics , Mutation , Spliceosomes/metabolism , Animals , Humans , RNA Splice Sites , RNA Splicing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spliceosomes/genetics
5.
Cell ; 161(6): 1474-e1, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046445

ABSTRACT

Spliceosomes are multi-megadalton RNA-protein molecular machines that carry out pre-mRNA splicing, that is, the removal of non-coding intervening sequences (introns) from eukaryotic pre-mRNAs and the ligation of neighboring coding regions (exons) to produce mature mRNA for protein biosynthesis on the ribosome. They are the prototypes of dynamic molecular machines, assembling de novo for each splicing event by the stepwise recruitment of subunits on a substrate.


Subject(s)
Disease/genetics , RNA Splicing , Spliceosomes/metabolism , Yeasts/metabolism , Animals , Humans , Yeasts/genetics
6.
Nature ; 630(8018): 1012-1019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778104

ABSTRACT

Early spliceosome assembly can occur through an intron-defined pathway, whereby U1 and U2 small nuclear ribonucleoprotein particles (snRNPs) assemble across the intron1. Alternatively, it can occur through an exon-defined pathway2-5, whereby U2 binds the branch site located upstream of the defined exon and U1 snRNP interacts with the 5' splice site located directly downstream of it. The U4/U6.U5 tri-snRNP subsequently binds to produce a cross-intron (CI) or cross-exon (CE) pre-B complex, which is then converted to the spliceosomal B complex6,7. Exon definition promotes the splicing of upstream introns2,8,9 and plays a key part in alternative splicing regulation10-16. However, the three-dimensional structure of exon-defined spliceosomal complexes and the molecular mechanism of the conversion from a CE-organized to a CI-organized spliceosome, a pre-requisite for splicing catalysis, remain poorly understood. Here cryo-electron microscopy analyses of human CE pre-B complex and B-like complexes reveal extensive structural similarities with their CI counterparts. The results indicate that the CE and CI spliceosome assembly pathways converge already at the pre-B stage. Add-back experiments using purified CE pre-B complexes, coupled with cryo-electron microscopy, elucidate the order of the extensive remodelling events that accompany the formation of B complexes and B-like complexes. The molecular triggers and roles of B-specific proteins in these rearrangements are also identified. We show that CE pre-B complexes can productively bind in trans to a U1 snRNP-bound 5' splice site. Together, our studies provide new mechanistic insights into the CE to CI switch during spliceosome assembly and its effect on pre-mRNA splice site pairing at this stage.


Subject(s)
Exons , Introns , RNA Splicing , Spliceosomes , Humans , Alternative Splicing , Cryoelectron Microscopy , Exons/genetics , Introns/genetics , Models, Molecular , RNA Splice Sites/genetics , RNA Splicing/genetics , Spliceosomes/metabolism , Spliceosomes/chemistry , Spliceosomes/ultrastructure , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/ultrastructure
7.
EMBO J ; 43(6): 1065-1088, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383864

ABSTRACT

The B complex is a key intermediate stage of spliceosome assembly. To improve the structural resolution of monomeric, human spliceosomal B (hB) complexes and thereby generate a more comprehensive hB molecular model, we determined the cryo-EM structure of B complex dimers formed in the presence of ATP γ S. The enhanced resolution of these complexes allows a finer molecular dissection of how the 5' splice site (5'ss) is recognized in hB, and new insights into molecular interactions of FBP21, SNU23 and PRP38 with the U6/5'ss helix and with each other. It also reveals that SMU1 and RED are present as a heterotetrameric complex and are located at the interface of the B dimer protomers. We further show that MFAP1 and UBL5 form a 5' exon binding channel in hB, and elucidate the molecular contacts stabilizing the 5' exon at this stage. Our studies thus yield more accurate models of protein and RNA components of hB complexes. They further allow the localization of additional proteins and protein domains (such as SF3B6, BUD31 and TCERG1) whose position was not previously known, thereby uncovering new functions for B-specific and other hB proteins during pre-mRNA splicing.


Subject(s)
RNA Splicing , Spliceosomes , Humans , Spliceosomes/genetics , Cryoelectron Microscopy , RNA Splice Sites , Exons , RNA Precursors/genetics , RNA Precursors/metabolism , Transcriptional Elongation Factors/genetics , Nuclear Proteins/metabolism
8.
Mol Cell ; 80(1): 127-139.e6, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33007253

ABSTRACT

Human spliceosomes contain numerous proteins absent in yeast, whose functions remain largely unknown. Here we report a 3D cryo-EM structure of the human spliceosomal C complex at 3.4 Å core resolution and 4.5-5.7 Å at its periphery, and aided by protein crosslinking we determine its molecular architecture. Our structure provides additional insights into the spliceosome's architecture between the catalytic steps of splicing, and how proteins aid formation of the spliceosome's catalytically active RNP (ribonucleoprotein) conformation. It reveals the spatial organization of the metazoan-specific proteins PPWD1, WDR70, FRG1, and CIR1 in human C complexes, indicating they stabilize functionally important protein domains and RNA structures rearranged/repositioned during the Bact to C transition. Structural comparisons with human Bact, C∗, and P complexes reveal an intricate cascade of RNP rearrangements during splicing catalysis, with intermediate RNP conformations not found in yeast, and additionally elucidate the structural basis for the sequential recruitment of metazoan-specific spliceosomal proteins.


Subject(s)
RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , Spliceosomes/metabolism , Animals , Catalysis , HeLa Cells , Humans , Introns/genetics , Models, Molecular , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Stability , RNA/chemistry , RNA/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae/metabolism , Species Specificity , Time Factors
9.
Mol Cell ; 77(6): 1322-1339.e11, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32006464

ABSTRACT

Deregulated expression of MYC induces a dependence on the NUAK1 kinase, but the molecular mechanisms underlying this dependence have not been fully clarified. Here, we show that NUAK1 is a predominantly nuclear protein that associates with a network of nuclear protein phosphatase 1 (PP1) interactors and that PNUTS, a nuclear regulatory subunit of PP1, is phosphorylated by NUAK1. Both NUAK1 and PNUTS associate with the splicing machinery. Inhibition of NUAK1 abolishes chromatin association of PNUTS, reduces spliceosome activity, and suppresses nascent RNA synthesis. Activation of MYC does not bypass the requirement for NUAK1 for spliceosome activity but significantly attenuates transcription inhibition. Consequently, NUAK1 inhibition in MYC-transformed cells induces global accumulation of RNAPII both at the pause site and at the first exon-intron boundary but does not increase mRNA synthesis. We suggest that NUAK1 inhibition in the presence of deregulated MYC traps non-productive RNAPII because of the absence of correctly assembled spliceosomes.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Protein Kinases/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Spliceosomes/metabolism , Transcription, Genetic , Animals , Cell Nucleus/genetics , Chromatin/genetics , Gene Expression Regulation , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Protein Kinases/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing , Repressor Proteins/genetics , Spliceosomes/genetics
10.
Nature ; 596(7871): 296-300, 2021 08.
Article in English | MEDLINE | ID: mdl-34349264

ABSTRACT

During the splicing of introns from precursor messenger RNAs (pre-mRNAs), the U2 small nuclear ribonucleoprotein (snRNP) must undergo stable integration into the spliceosomal A complex-a poorly understood, multistep process that is facilitated by the DEAD-box helicase Prp5 (refs. 1-4). During this process, the U2 small nuclear RNA (snRNA) forms an RNA duplex with the pre-mRNA branch site (the U2-BS helix), which is proofread by Prp5 at this stage through an unclear mechanism5. Here, by deleting the branch-site adenosine (BS-A) or mutating the branch-site sequence of an actin pre-mRNA, we stall the assembly of spliceosomes in extracts from the yeast Saccharomyces cerevisiae directly before the A complex is formed. We then determine the three-dimensional structure of this newly identified assembly intermediate by cryo-electron microscopy. Our structure indicates that the U2-BS helix has formed in this pre-A complex, but is not yet clamped by the HEAT domain of the Hsh155 protein (Hsh155HEAT), which exhibits an open conformation. The structure further reveals a large-scale remodelling/repositioning of the U1 and U2 snRNPs during the formation of the A complex that is required to allow subsequent binding of the U4/U6.U5 tri-snRNP, but that this repositioning is blocked in the pre-A complex by the presence of Prp5. Our data suggest that binding of Hsh155HEAT to the bulged BS-A of the U2-BS helix triggers closure of Hsh155HEAT, which in turn destabilizes Prp5 binding. Thus, Prp5 proofreads the branch site indirectly, hindering spliceosome assembly if branch-site mutations prevent the remodelling of Hsh155HEAT. Our data provide structural insights into how a spliceosomal helicase enhances the fidelity of pre-mRNA splicing.


Subject(s)
DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Splicing , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Spliceosomes/enzymology , Actins/genetics , Adenosine/metabolism , Binding Sites , Cryoelectron Microscopy , DEAD-box RNA Helicases/ultrastructure , Models, Molecular , Mutation , Protein Domains , RNA Precursors/metabolism , RNA Precursors/ultrastructure , RNA Splicing/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Spliceosomes/chemistry , Spliceosomes/metabolism
11.
Nature ; 583(7815): 310-313, 2020 07.
Article in English | MEDLINE | ID: mdl-32494006

ABSTRACT

The U2 small nuclear ribonucleoprotein (snRNP) has an essential role in the selection of the precursor mRNA branch-site adenosine, the nucleophile for the first step of splicing1. Stable addition of U2 during early spliceosome formation requires the DEAD-box ATPase PRP52-7. Yeast U2 small nuclear RNA (snRNA) nucleotides that form base pairs with the branch site are initially sequestered in a branchpoint-interacting stem-loop (BSL)8, but whether the human U2 snRNA folds in a similar manner is unknown. The U2 SF3B1 protein, a common mutational target in haematopoietic cancers9, contains a HEAT domain (SF3B1HEAT) with an open conformation in isolated SF3b10, but a closed conformation in spliceosomes11, which is required for stable interaction between U2 and the branch site. Here we report a 3D cryo-electron microscopy structure of the human 17S U2 snRNP at a core resolution of 4.1 Å and combine it with protein crosslinking data to determine the molecular architecture of this snRNP. Our structure reveals that SF3B1HEAT interacts with PRP5 and TAT-SF1, and maintains its open conformation in U2 snRNP, and that U2 snRNA forms a BSL that is sandwiched between PRP5, TAT-SF1 and SF3B1HEAT. Thus, substantial remodelling of the BSL and displacement of BSL-interacting proteins must occur to allow formation of the U2-branch-site helix. Our studies provide a structural explanation of why TAT-SF1 must be displaced before the stable addition of U2 to the spliceosome, and identify RNP rearrangements facilitated by PRP5 that are required for stable interaction between U2 and the branch site.


Subject(s)
Cryoelectron Microscopy , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/ultrastructure , Base Sequence , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , HeLa Cells , Humans , Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Conformation , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism
12.
Mol Cell ; 70(2): 265-273.e8, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656923

ABSTRACT

SF3B is a multi-protein complex essential for branch site (BS) recognition and selection during pre-mRNA splicing. Several splicing modulators with antitumor activity bind SF3B and thereby modulate splicing. Here we report the crystal structure of a human SF3B core in complex with pladienolide B (PB), a macrocyclic splicing modulator and potent inhibitor of tumor cell proliferation. PB stalls SF3B in an open conformation by acting like a wedge within a hinge, modulating SF3B's transition to the closed conformation needed to form the BS adenosine-binding pocket and stably accommodate the BS/U2 duplex. This work explains the structural basis for the splicing modulation activity of PB and related compounds, and reveals key interactions between SF3B and a common pharmacophore, providing a framework for future structure-based drug design.


Subject(s)
Antineoplastic Agents/pharmacology , Epoxy Compounds/pharmacology , Macrolides/pharmacology , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , RNA Splicing/drug effects , Adenosine/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Drug Design , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , HCT116 Cells , HeLa Cells , Humans , Macrolides/chemistry , Macrolides/metabolism , Models, Molecular , Multiprotein Complexes , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Conformation , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Sf9 Cells , Structure-Activity Relationship , Trans-Activators
13.
Mol Cell ; 69(6): 979-992.e6, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29547724

ABSTRACT

Human nineteen complex (NTC) acts as a multimeric E3 ubiquitin ligase in DNA repair and splicing. The transfer of ubiquitin is mediated by Prp19-a homotetrameric component of NTC whose elongated coiled coils serve as an assembly axis for two other proteins called SPF27 and CDC5L. We find that Prp19 is inactive on its own and have elucidated the structural basis of its autoinhibition by crystallography and mutational analysis. Formation of the NTC core by stepwise assembly of SPF27, CDC5L, and PLRG1 onto the Prp19 tetramer enables ubiquitin ligation. Protein-protein crosslinking of NTC, functional assays in vitro, and assessment of its role in DNA damage response provide mechanistic insight into the organization of the NTC core and the communication between PLRG1 and Prp19 that enables E3 activity. This reveals a unique mode of regulation for a complex E3 ligase and advances understanding of its dynamics in various cellular pathways.


Subject(s)
DNA Repair Enzymes/metabolism , Nuclear Proteins/metabolism , RNA Splicing Factors/metabolism , Animals , Cell Cycle Proteins/metabolism , Crystallization , DNA Damage , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Mutation , Neoplasm Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Conformation , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA-Binding Proteins/metabolism , Replication Protein A/metabolism , Sf9 Cells , Spodoptera , Structure-Activity Relationship , Ubiquitination , WD40 Repeats
14.
Genes Dev ; 31(23-24): 2416-2429, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29330354

ABSTRACT

The precise function of the trimeric retention and splicing (RES) complex in pre-mRNA splicing remains unclear. Here we dissected the role of RES during the assembly and activation of yeast spliceosomes. The efficiency of pre-mRNA splicing was significantly lower in the absence of the RES protein Snu17, and the recruitment of its binding partners, Pml1 (pre-mRNA leakage protein 1) and Bud13 (bud site selection protein 13), to the spliceosome was either abolished or substantially reduced. RES was not required for the assembly of spliceosomal B complexes, but its absence hindered efficient Bact complex formation. ΔRES spliceosomes were no longer strictly dependent on Prp2 activity for their catalytic activation, suggesting that they are structurally compromised. Addition of Prp2, Spp2, and UTP to affinity-purified ΔRES B or a mixture of B/Bact complexes formed on wild-type pre-mRNA led to their disassembly. However, no substantial disassembly was observed with ΔRES spliceosomes formed on a truncated pre-mRNA that allows Prp2 binding but blocks its activity. Thus, in the absence of RES, Prp2 appears to bind prematurely, leading to the disassembly of the ΔRES B complexes to which it binds. Our data suggest that Prp2 can dismantle B complexes with an aberrant protein composition, suggesting that it may proofread the spliceosome's RNP structure prior to activation.


Subject(s)
RNA Splicing/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , DEAD-box RNA Helicases/metabolism , Protein Multimerization/genetics , RNA Precursors/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Spliceosomes/genetics
15.
Cell ; 136(4): 701-18, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19239890

ABSTRACT

Ribonucleoproteins (RNPs) mediate key cellular functions such as gene expression and its regulation. Whereas most RNP enzymes are stable in composition and harbor preformed active sites, the spliceosome, which removes noncoding introns from precursor messenger RNAs (pre-mRNAs), follows fundamentally different strategies. In order to provide both accuracy to the recognition of reactive splice sites in the pre-mRNA and flexibility to the choice of splice sites during alternative splicing, the spliceosome exhibits exceptional compositional and structural dynamics that are exploited during substrate-dependent complex assembly, catalytic activation, and active site remodeling.


Subject(s)
RNA Splicing , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Spliceosomes/chemistry , Spliceosomes/metabolism , Animals , Humans , RNA Splice Sites
16.
Mol Cell ; 64(2): 307-319, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27720643

ABSTRACT

SF3b is a heptameric protein complex of the U2 small nuclear ribonucleoprotein (snRNP) that is essential for pre-mRNA splicing. Mutations in the largest SF3b subunit, SF3B1/SF3b155, are linked to cancer and lead to alternative branch site (BS) selection. Here we report the crystal structure of a human SF3b core complex, revealing how the distinctive conformation of SF3b155's HEAT domain is maintained by multiple contacts with SF3b130, SF3b10, and SF3b14b. Protein-protein crosslinking enabled the localization of the BS-binding proteins p14 and U2AF65 within SF3b155's HEAT-repeat superhelix, which together with SF3b14b forms a composite RNA-binding platform. SF3b155 residues, the mutation of which leads to cancer, contribute to the tertiary structure of the HEAT superhelix and its surface properties in the proximity of p14 and U2AF65. The molecular architecture of SF3b reveals the spatial organization of cancer-related SF3b155 mutations and advances our understanding of their effects on SF3b structure and function.


Subject(s)
Mutation , Neoplasm Proteins/chemistry , Oncogene Proteins/chemistry , Phosphoproteins/chemistry , RNA Splicing Factors/chemistry , Spliceosomes/chemistry , Splicing Factor U2AF/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Genes, Tumor Suppressor , HeLa Cells , Humans , Models, Molecular , Moths , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spliceosomes/metabolism , Spliceosomes/ultrastructure , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
18.
Cell ; 135(7): 1224-36, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109894

ABSTRACT

Alternative splicing makes a major contribution to proteomic diversity in higher eukaryotes with approximately 70% of genes encoding two or more isoforms. In most cases, the molecular mechanisms responsible for splice site choice remain poorly understood. Here, we used a randomization-selection approach in vitro to identify sequence elements that could silence a proximal strong 5' splice site located downstream of a weakened 5' splice site. We recovered two exonic and four intronic motifs that effectively silenced the proximal 5' splice site both in vitro and in vivo. Surprisingly, silencing was only observed in the presence of the competing upstream 5' splice site. Biochemical evidence strongly suggests that the silencing motifs function by altering the U1 snRNP/5' splice site complex in a manner that impairs commitment to specific splice site pairing. The data indicate that perturbations of non-rate-limiting step(s) in splicing can lead to dramatic shifts in splice site choice.


Subject(s)
Alternative Splicing , Gene Expression Regulation , RNA Splice Sites , Exons , Genetic Techniques , HeLa Cells , Humans , Models, Biological
19.
Nature ; 542(7641): 318-323, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28076346

ABSTRACT

Spliceosome rearrangements facilitated by RNA helicase PRP16 before catalytic step two of splicing are poorly understood. Here we report a 3D cryo-electron microscopy structure of the human spliceosomal C complex stalled directly after PRP16 action (C*). The architecture of the catalytic U2-U6 ribonucleoprotein (RNP) core of the human C* spliceosome is very similar to that of the yeast pre-Prp16 C complex. However, in C* the branched intron region is separated from the catalytic centre by approximately 20 Å, and its position close to the U6 small nuclear RNA ACAGA box is stabilized by interactions with the PRP8 RNase H-like and PRP17 WD40 domains. RNA helicase PRP22 is located about 100 Å from the catalytic centre, suggesting that it destabilizes the spliced mRNA after step two from a distance. Comparison of the structure of the yeast C and human C* complexes reveals numerous RNP rearrangements that are likely to be facilitated by PRP16, including a large-scale movement of the U2 small nuclear RNP.


Subject(s)
Cryoelectron Microscopy , RNA Splicing , Spliceosomes/metabolism , Spliceosomes/ultrastructure , Adenosine/metabolism , Base Sequence , Biocatalysis , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/ultrastructure , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/ultrastructure , Exons/genetics , Humans , Introns/genetics , Models, Molecular , Movement , Protein Domains , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA Splicing Factors/ultrastructure , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/ultrastructure , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/ultrastructure , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Spliceosomes/chemistry
20.
Nucleic Acids Res ; 49(10): 5845-5866, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34023904

ABSTRACT

Splicing is catalyzed by the spliceosome, a compositionally dynamic complex assembled stepwise on pre-mRNA. We reveal links between splicing machinery components and the intrinsically disordered ciliopathy protein SANS. Pathogenic mutations in SANS/USH1G lead to Usher syndrome-the most common cause of deaf-blindness. Previously, SANS was shown to function only in the cytosol and primary cilia. Here, we have uncovered molecular links between SANS and pre-mRNA splicing catalyzed by the spliceosome in the nucleus. We show that SANS is found in Cajal bodies and nuclear speckles, where it interacts with components of spliceosomal sub-complexes such as SF3B1 and the large splicing cofactor SON but also with PRPFs and snRNAs related to the tri-snRNP complex. SANS is required for the transfer of tri-snRNPs between Cajal bodies and nuclear speckles for spliceosome assembly and may also participate in snRNP recycling back to Cajal bodies. SANS depletion alters the kinetics of spliceosome assembly, leading to accumulation of complex A. SANS deficiency and USH1G pathogenic mutations affects splicing of genes related to cell proliferation and human Usher syndrome. Thus, we provide the first evidence that splicing dysregulation may participate in the pathophysiology of Usher syndrome.


Subject(s)
Alternative Splicing/genetics , Nerve Tissue Proteins/metabolism , RNA Precursors/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Usher Syndromes/metabolism , Cell Nucleus/metabolism , Cell Proliferation/genetics , Coiled Bodies/metabolism , DNA-Binding Proteins/metabolism , Eye Proteins/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mass Spectrometry , Microscopy, Electron, Transmission , Minor Histocompatibility Antigens/metabolism , Nerve Tissue Proteins/genetics , Phosphoproteins/metabolism , Proteomics , RNA Precursors/genetics , RNA Splicing Factors/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Spliceosomes/genetics , Transcription Factors/metabolism , Usher Syndromes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL