Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cell ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38823386

ABSTRACT

Integrator is a multi-subunit protein complex responsible for premature transcription termination of coding and non-coding RNAs. This is achieved via two enzymatic activities, RNA endonuclease and protein phosphatase, acting on the promoter-proximally paused RNA polymerase Ⅱ (RNAPⅡ). Yet, it remains unclear how Integrator assembly and recruitment are regulated and what the functions of many of its core subunits are. Here, we report the structures of two human Integrator sub-complexes: INTS10/13/14/15 and INTS5/8/10/15, and an integrative model of the fully assembled Integrator bound to the RNAPⅡ paused elongating complex (PEC). An in silico protein-protein interaction screen of over 1,500 human transcription factors (TFs) identified ZNF655 as a direct interacting partner of INTS13 within the fully assembled Integrator. We propose a model wherein INTS13 acts as a platform for the recruitment of TFs that could modulate the stability of the Integrator's association at specific loci and regulate transcription attenuation of the target genes.

2.
Nat Struct Mol Biol ; 31(5): 752-756, 2024 May.
Article in English | MEDLINE | ID: mdl-38467877

ABSTRACT

The 20S U5 small nuclear ribonucleoprotein particle (snRNP) is a 17-subunit RNA-protein complex and a precursor of the U4/U6.U5 tri-snRNP, the major building block of the precatalytic spliceosome. CD2BP2 is a hallmark protein of the 20S U5 snRNP, absent from the mature tri-snRNP. Here we report a high-resolution cryogenic electron microscopy structure of the 20S U5 snRNP, shedding light on the mutually exclusive interfaces utilized during tri-snRNP assembly and the role of the CD2BP2 in facilitating this process.


Subject(s)
Cryoelectron Microscopy , Models, Molecular , Ribonucleoprotein, U5 Small Nuclear , Humans , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Spliceosomes/metabolism , Spliceosomes/chemistry , Spliceosomes/ultrastructure , Protein Conformation , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry
3.
Nat Commun ; 14(1): 8482, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38123540

ABSTRACT

Cleavage and formation of phosphodiester bonds in nucleic acids is accomplished by large cellular machineries composed of both protein and RNA. Long thought to rely on a two-metal-ion mechanism for catalysis, structure comparisons revealed many contain highly spatially conserved second-shell monovalent cations, whose precise function remains elusive. A recent high-resolution structure of the spliceosome, essential for pre-mRNA splicing in eukaryotes, revealed a potassium ion in the active site. Here, we employ biased quantum mechanics/ molecular mechanics molecular dynamics to elucidate the function of this monovalent ion in splicing. We discover that the K+ ion regulates the kinetics and thermodynamics of the first splicing step by rigidifying the active site and stabilizing the substrate in the pre- and post-catalytic state via formation of key hydrogen bonds. Our work supports a direct role for the K+ ion during catalysis and provides a mechanistic hypothesis likely shared by other nucleic acid processing enzymes.


Subject(s)
RNA , Spliceosomes , Spliceosomes/metabolism , RNA/metabolism , RNA Splicing , Catalysis , Metals/metabolism , Potassium/metabolism , Chelating Agents/metabolism , Nucleic Acid Conformation , Binding Sites , Cations, Monovalent/metabolism
4.
Genes Dev ; 37(21-24): 968-983, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-37977822

ABSTRACT

The spliceosomal gene SF3B1 is frequently mutated in cancer. While it is known that SF3B1 hotspot mutations lead to loss of splicing factor SUGP1 from spliceosomes, the cancer-relevant SF3B1-SUGP1 interaction has not been characterized. To address this issue, we show by structural modeling that two regions flanking the SUGP1 G-patch make numerous contacts with the region of SF3B1 harboring hotspot mutations. Experiments confirmed that all the cancer-associated mutations in these regions, as well as mutations affecting other residues in the SF3B1-SUGP1 interface, not only weaken or disrupt the interaction but also alter splicing similarly to SF3B1 cancer mutations. Finally, structural modeling of a trimeric protein complex reveals that the SF3B1-SUGP1 interaction "loops out" the G-patch for interaction with the helicase DHX15. Our study thus provides an unprecedented molecular view of a protein complex essential for accurate splicing and also reveals that numerous cancer-associated mutations disrupt the critical SF3B1-SUGP1 interaction.


Subject(s)
Neoplasms , Spliceosomes , Humans , RNA, Messenger/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , RNA Splicing Factors/chemistry , RNA Splicing/genetics , Neoplasms/genetics , Neoplasms/metabolism , Mutation , Phosphoproteins/metabolism
5.
Cell Rep ; 42(3): 112244, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36920904

ABSTRACT

RNA polymerase II (RNAPII) controls expression of all protein-coding genes and most noncoding loci in higher eukaryotes. Calibrating RNAPII activity requires an assortment of polymerase-associated factors that are recruited at sites of active transcription. The Integrator complex is one of the most elusive transcriptional regulators in metazoans, deemed to be recruited after initiation to help establish and modulate paused RNAPII. Integrator is known to be composed of 14 subunits that assemble and operate in a modular fashion. We employed proteomics and machine-learning structure prediction (AlphaFold2) to identify an additional Integrator subunit, INTS15. We report that INTS15 assembles primarily with the INTS13/14/10 module and interfaces with the Int-PP2A module. Functional genomics analysis further reveals a role for INTS15 in modulating RNAPII pausing at a subset of genes. Our study shows that omics approaches combined with AlphaFold2-based predictions provide additional insights into the molecular architecture of large and dynamic multiprotein complexes.


Subject(s)
RNA Polymerase II , Transcription, Genetic , RNA Polymerase II/metabolism
6.
Mol Cell ; 82(13): 2355-2357, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35803215

ABSTRACT

Two recent studies by Gerlach et al. (2022) and Puno and Lima (2022) provide new structural and functional insight into the assembly of the nuclear exosome targeting complex (NEXT) and how it may target specific classes of RNA for degradation.


Subject(s)
Exosomes , Cell Nucleus/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/metabolism , RNA/metabolism
7.
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
8.
Mol Cell ; 81(7): 1439-1452.e9, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33705709

ABSTRACT

The ATPase Prp16 governs equilibrium between the branching (B∗/C) and exon ligation (C∗/P) conformations of the spliceosome. Here, we present the electron cryomicroscopy reconstruction of the Saccharomyces cerevisiae C-complex spliceosome at 2.8 Å resolution and identify a novel C-complex intermediate (Ci) that elucidates the molecular basis for this equilibrium. The exon-ligation factors Prp18 and Slu7 bind to Ci before ATP hydrolysis by Prp16 can destabilize the branching conformation. Biochemical assays suggest that these pre-bound factors prime the C complex for conversion to C∗ by Prp16. A complete model of the Prp19 complex (NTC) reveals how the branching factors Yju2 and Isy1 are recruited by the NTC before branching. Prp16 remodels Yju2 binding after branching, allowing Yju2 to remain tethered to the NTC in the C∗ complex to promote exon ligation. Our results explain how Prp16 action modulates the dynamic binding of step-specific factors to alternatively stabilize the C or C∗ conformation and establish equilibrium of the catalytic spliceosome.


Subject(s)
Models, Chemical , RNA Splicing , RNA, Fungal/chemistry , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Spliceosomes/chemistry , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism
9.
Mol Cell ; 81(6): 1246-1259.e8, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33548203

ABSTRACT

The Integrator is a specialized 3' end-processing complex involved in cleavage and transcription termination of a subset of nascent RNA polymerase II transcripts, including small nuclear RNAs (snRNAs). We provide evidence of the modular nature of the Integrator complex by biochemically characterizing its two subcomplexes, INTS5/8 and INTS10/13/14. Using cryoelectron microscopy (cryo-EM), we determined a 3.5-Å-resolution structure of the INTS4/9/11 ternary complex, which constitutes Integrator's catalytic core. Our structure reveals the spatial organization of the catalytic nuclease INTS11, bound to its catalytically impaired homolog INTS9 via several interdependent interfaces. INTS4, a helical repeat protein, plays a key role in stabilizing nuclease domains and other components. In this assembly, all three proteins form a composite electropositive groove, suggesting a putative RNA binding path within the complex. Comparison with other 3' end-processing machineries points to distinct features and a unique architecture of the Integrator's catalytic module.


Subject(s)
Multiprotein Complexes , Transcription Termination, Genetic , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
10.
Transcription ; 12(5): 251-265, 2021 10.
Article in English | MEDLINE | ID: mdl-35311473

ABSTRACT

The Integrator was originally discovered as a specialized 3'-end processing endonuclease complex required for maturation of RNA polymerase II (RNAPII)-dependent small nuclear RNAs (snRNAs). Since its discovery, Integrator's spectrum of substrates was significantly expanded to include non-polyadenylated long noncoding RNAs (lncRNA), enhancer RNAs (eRNAs), telomerase RNA (tertRNA), several Herpesvirus transcripts, and messenger RNAs (mRNAs). Recently emerging transcriptome-wide studies reveled an important role of the Integrator in protein-coding genes, where it contributes to gene expression regulation through promoter-proximal transcription attenuation. These new functional data are complemented by several structures of Integrator modules and higher-order complexes, providing mechanistic insights into Integrator-mediated processing events. In this work, we summarize recent progress in our understanding of the structure and function of the Integrator complex.


Subject(s)
RNA Polymerase II , RNA, Small Nuclear , Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics
11.
Nature ; 572(7769): 382-386, 2019 08.
Article in English | MEDLINE | ID: mdl-31330532

ABSTRACT

The family of bacterial SidE enzymes catalyses phosphoribosyl-linked serine ubiquitination and promotes infectivity of Legionella pneumophila, a pathogenic bacteria that causes Legionnaires' disease1-3. SidE enzymes share the genetic locus with the Legionella effector SidJ that spatiotemporally opposes the toxicity of these enzymes in yeast and mammalian cells, through a mechanism that is currently unknown4-6. Deletion of SidJ leads to a substantial defect in the growth of Legionella in both its natural hosts (amoebae) and in mouse macrophages4,5. Here we demonstrate that SidJ is a glutamylase that modifies the catalytic glutamate in the mono-ADP ribosyl transferase domain of the SdeA, thus blocking the ubiquitin ligase activity of SdeA. The glutamylation activity of SidJ requires interaction with the eukaryotic-specific co-factor calmodulin, and can be regulated by intracellular changes in Ca2+ concentrations. The cryo-electron microscopy structure of SidJ in complex with human apo-calmodulin revealed the architecture of this heterodimeric glutamylase. We show that, in cells infected with L. pneumophila, SidJ mediates the glutamylation of SidE enzymes on the surface of vacuoles that contain Legionella. We used quantitative proteomics to uncover multiple host proteins as putative targets of SidJ-mediated glutamylation. Our study reveals the mechanism by which SidE ligases are inhibited by a SidJ-calmodulin glutamylase, and opens avenues for exploring an understudied protein modification (glutamylation) in eukaryotes.


Subject(s)
Bacterial Proteins/metabolism , Calmodulin/metabolism , Glutamic Acid/metabolism , Legionella pneumophila/enzymology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin/metabolism , Virulence Factors/metabolism , ADP-Ribosylation , Apoproteins/metabolism , Bacterial Proteins/agonists , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Calmodulin/pharmacology , Catalysis , Cryoelectron Microscopy , Crystallography, X-Ray , HEK293 Cells , Humans , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Virulence Factors/agonists , Virulence Factors/chemistry
12.
Biochem Soc Trans ; 46(6): 1407-1422, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30420411

ABSTRACT

The spliceosome is a multi-subunit RNA-protein complex involved in the removal of non-coding segments (introns) from between the coding regions (exons) in precursors of messenger RNAs (pre-mRNAs). Intron removal proceeds via two transesterification reactions, occurring between conserved sequences at intron-exon junctions. A tightly regulated, hierarchical assembly with a multitude of structural and compositional rearrangements posed a great challenge for structural studies of the spliceosome. Over the years, X-ray crystallography dominated the field, providing valuable high-resolution structural information that was mostly limited to individual proteins and smaller sub-complexes. Recent developments in the field of cryo-electron microscopy allowed the visualisation of fully assembled yeast and human spliceosomes, providing unprecedented insights into substrate recognition, catalysis, and active site formation. This has advanced our mechanistic understanding of pre-mRNA splicing enormously.


Subject(s)
RNA Precursors/metabolism , Spliceosomes/metabolism , Catalysis , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Introns/genetics , Introns/physiology , RNA Precursors/genetics , Spliceosomes/genetics , Spliceosomes/ultrastructure
13.
Chem Rev ; 118(8): 4156-4176, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29377672

ABSTRACT

Nuclear pre-mRNA splicing and group II intron self-splicing both proceed by two-step transesterification reactions via a lariat intron intermediate. Recently determined cryo-electron microscopy (cryo-EM) structures of catalytically active spliceosomes revealed the RNA-based catalytic core and showed how pre-mRNA substrates and reaction products are positioned in the active site. These findings highlight a strong structural similarity to the group II intron active site, strengthening the notion that group II introns and spliceosomes evolved from a common ancestor. Prp8, the largest and most conserved protein in the spliceosome, cradles the active site RNA. Prp8 and group II intron maturase have a similar domain architecture, suggesting that they also share a common evolutionary origin. The interactions between maturase and key group II intron RNA elements, such as the exon-binding loop and domains V and VI, are recapitulated in the interactions between Prp8 and key elements in the spliceosome's catalytic RNA core. Structural comparisons suggest that the extensive RNA scaffold of the group II intron was gradually replaced by proteins as the spliceosome evolved. A plausible model of spliceosome evolution is discussed.


Subject(s)
Cryoelectron Microscopy/methods , Introns , Nucleic Acid Conformation , RNA Precursors/chemistry , RNA Splicing , RNA, Messenger/chemistry , Cell Nucleus/chemistry , Crystallography, X-Ray , Exons , Hydrolysis , Phylogeny , RNA Precursors/ultrastructure , RNA, Messenger/ultrastructure , Spliceosomes
14.
Science ; 358(6368): 1283-1288, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29146871

ABSTRACT

Introns are removed from eukaryotic messenger RNA precursors by the spliceosome in two transesterification reactions-branching and exon ligation. The mechanism of 3'-splice site recognition during exon ligation has remained unclear. Here we present the 3.7-angstrom cryo-electron microscopy structure of the yeast P-complex spliceosome immediately after exon ligation. The 3'-splice site AG dinucleotide is recognized through non-Watson-Crick pairing with the 5' splice site and the branch-point adenosine. After the branching reaction, protein factors work together to remodel the spliceosome and stabilize a conformation competent for 3'-splice site docking, thereby promoting exon ligation. The structure accounts for the strict conservation of the GU and AG dinucleotides at the 5' and 3' ends of introns and provides insight into the catalytic mechanism of exon ligation.


Subject(s)
Exons/genetics , RNA Splice Sites , RNA Splicing , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Spliceosomes/chemistry , Base Pairing , Catalytic Domain , Cryoelectron Microscopy , Introns/genetics , Protein Conformation , RNA Precursors/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Spliceosomes/ultrastructure
15.
Nature ; 542(7641): 377-380, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28076345

ABSTRACT

The spliceosome excises introns from pre-mRNAs in two sequential transesterifications-branching and exon ligation-catalysed at a single catalytic metal site in U6 small nuclear RNA (snRNA). Recently reported structures of the spliceosomal C complex with the cleaved 5' exon and lariat-3'-exon bound to the catalytic centre revealed that branching-specific factors such as Cwc25 lock the branch helix into position for nucleophilic attack of the branch adenosine at the 5' splice site. Furthermore, the ATPase Prp16 is positioned to bind and translocate the intron downstream of the branch point to destabilize branching-specific factors and release the branch helix from the active site. Here we present, at 3.8 Å resolution, the cryo-electron microscopy structure of a Saccharomyces cerevisiae spliceosome stalled after Prp16-mediated remodelling but before exon ligation. While the U6 snRNA catalytic core remains firmly held in the active site cavity of Prp8 by proteins common to both steps, the branch helix has rotated by 75° compared to the C complex and is stabilized in a new position by Prp17, Cef1 and the reoriented Prp8 RNase H-like domain. This rotation of the branch helix removes the branch adenosine from the catalytic core, creates a space for 3' exon docking, and restructures the pairing of the 5' splice site with the U6 snRNA ACAGAGA region. Slu7 and Prp18, which promote exon ligation, bind together to the Prp8 RNase H-like domain. The ATPase Prp22, bound to Prp8 in place of Prp16, could interact with the 3' exon, suggesting a possible basis for mRNA release after exon ligation. Together with the structure of the C complex, our structure of the C* complex reveals the two major conformations of the spliceosome during the catalytic stages of splicing.


Subject(s)
Cryoelectron Microscopy , Exons , RNA Splicing , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , Spliceosomes/ultrastructure , Adenosine/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/ultrastructure , Biocatalysis , Catalytic Domain , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/ultrastructure , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/ultrastructure , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Exons/genetics , Protein Binding , Protein Domains , RNA Helicases/metabolism , RNA Helicases/ultrastructure , RNA Splice Sites/genetics , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA Splicing Factors/ultrastructure , RNA, Small Nuclear/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/ultrastructure , Ribonuclease H/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/ultrastructure , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/ultrastructure , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Spliceosomes/chemistry
16.
Nature ; 537(7619): 197-201, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27459055

ABSTRACT

Precursor mRNA (pre-mRNA) splicing proceeds by two consecutive transesterification reactions via a lariat-intron intermediate. Here we present the 3.8 Å cryo-electron microscopy structure of the spliceosome immediately after lariat formation. The 5'-splice site is cleaved but remains close to the catalytic Mg2+ site in the U2/U6 small nuclear RNA (snRNA) triplex, and the 5'-phosphate of the intron nucleotide G(+1) is linked to the branch adenosine 2'OH. The 5'-exon is held between the Prp8 amino-terminal and linker domains, and base-pairs with U5 snRNA loop 1. Non-Watson-Crick interactions between the branch helix and 5'-splice site dock the branch adenosine into the active site, while intron nucleotides +3 to +6 base-pair with the U6 snRNA ACAGAGA sequence. Isy1 and the step-one factors Yju2 and Cwc25 stabilize docking of the branch helix. The intron downstream of the branch site emerges between the Prp8 reverse transcriptase and linker domains and extends towards the Prp16 helicase, suggesting a plausible mechanism of remodelling before exon ligation.


Subject(s)
Cryoelectron Microscopy , RNA Precursors/metabolism , RNA Splicing , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Spliceosomes/metabolism , Spliceosomes/ultrastructure , Adenosine/metabolism , Adenosine Triphosphatases/metabolism , Base Pairing , Base Sequence , Catalytic Domain , Esterification , Exons/genetics , Introns/genetics , Magnesium/metabolism , Models, Molecular , Nuclear Proteins/metabolism , RNA Helicases/metabolism , RNA Precursors/chemistry , RNA Precursors/ultrastructure , RNA Splice Sites/genetics , RNA Splicing Factors/metabolism , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Fungal/ultrastructure , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Spliceosomes/chemistry
17.
Nature ; 530(7590): 298-302, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26829225

ABSTRACT

U4/U6.U5 tri-snRNP represents a substantial part of the spliceosome before activation. A cryo-electron microscopy structure of Saccharomyces cerevisiae U4/U6.U5 tri-snRNP at 3.7 Šresolution led to an essentially complete atomic model comprising 30 proteins plus U4/U6 and U5 small nuclear RNAs (snRNAs). The structure reveals striking interweaving interactions of the protein and RNA components, including extended polypeptides penetrating into subunit interfaces. The invariant ACAGAGA sequence of U6 snRNA, which base-pairs with the 5'-splice site during catalytic activation, forms a hairpin stabilized by Dib1 and Prp8 while the adjacent nucleotides interact with the exon binding loop 1 of U5 snRNA. Snu114 harbours GTP, but its putative catalytic histidine is held away from the γ-phosphate by hydrogen bonding to a tyrosine in the amino-terminal domain of Prp8. Mutation of this histidine to alanine has no detectable effect on yeast growth. The structure provides important new insights into the spliceosome activation process leading to the formation of the catalytic centre.


Subject(s)
Cryoelectron Microscopy , Ribonucleoproteins, Small Nuclear/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/ultrastructure , Base Pairing , Catalytic Domain , DNA Helicases/metabolism , Exons/genetics , Guanosine Triphosphate/metabolism , Hydrogen Bonding , Models, Molecular , Nucleic Acid Conformation , RNA Splice Sites , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism
18.
Curr Opin Struct Biol ; 36: 48-57, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26803803

ABSTRACT

The spliceosome is formed on pre-mRNA substrates from five small nuclear ribonucleoprotein particles (U1, U2, U4/U6 and U5 snRNPs), and numerous non-snRNP factors. Saccharomyces cerevisiae U4/U6.U5 tri-snRNP comprises U5 snRNA, U4/U6 snRNA duplex and approximately 30 proteins and represents a substantial part of the spliceosome before activation. Schizosaccharomyces pombe U2.U6.U5 spliceosomal complex is a post-catalytic intron lariat spliceosome containing U2 and U5 snRNPs, NTC (nineteen complex), NTC-related proteins (NTR), U6 snRNA, and an RNA intron lariat. Two recent papers describe near-complete atomic structures of these complexes based on cryoEM single-particle analysis. The U4/U6.U5 tri-snRNP structure provides crucial insight into the activation mechanism of the spliceosome. The U2.U6.U5 complex reveals the striking architecture of NTC and NTR and important features of the group II intron-like catalytic RNA core remaining after spliced mRNA is released. These two structures greatly advance our understanding of the mechanism of pre-mRNA splicing.


Subject(s)
Cryoelectron Microscopy , Macromolecular Substances/chemistry , Macromolecular Substances/ultrastructure , Spliceosomes/chemistry , Spliceosomes/ultrastructure , Animals , Humans , Nucleic Acid Conformation , Protein Binding , Protein Conformation , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Schizosaccharomyces/metabolism
19.
Nature ; 523(7558): 47-52, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26106855

ABSTRACT

U4/U6.U5 tri-snRNP is a 1.5-megadalton pre-assembled spliceosomal complex comprising U5 small nuclear RNA (snRNA), extensively base-paired U4/U6 snRNAs and more than 30 proteins, including the key components Prp8, Brr2 and Snu114. The tri-snRNP combines with a precursor messenger RNA substrate bound to U1 and U2 small nuclear ribonucleoprotein particles (snRNPs), and transforms into a catalytically active spliceosome after extensive compositional and conformational changes triggered by unwinding of the U4 and U6 (U4/U6) snRNAs. Here we use cryo-electron microscopy single-particle reconstruction of Saccharomyces cerevisiae tri-snRNP at 5.9 Å resolution to reveal the essentially complete organization of its RNA and protein components. The single-stranded region of U4 snRNA between its 3' stem-loop and the U4/U6 snRNA stem I is loaded into the Brr2 helicase active site ready for unwinding. Snu114 and the amino-terminal domain of Prp8 position U5 snRNA to insert its loop I, which aligns the exons for splicing, into the Prp8 active site cavity. The structure provides crucial insights into the activation process and the active site of the spliceosome.


Subject(s)
Models, Molecular , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Saccharomyces cerevisiae/chemistry , Spliceosomes/physiology , Binding Sites , Cryoelectron Microscopy , Protein Structure, Quaternary , RNA Helicases/chemistry , RNA Helicases/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/chemistry
20.
Curr Opin Struct Biol ; 25: 57-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24480332

ABSTRACT

Spliceosomes are large, dynamic ribonucleoprotein complexes that catalyse the removal of introns from messenger RNA precursors via a two-step splicing reaction. The recent crystal structure of Prp8 has revealed Reverse Transcriptase-like, Linker and Endonuclease-like domains. The intron branch-point cross-link with the Linker domain of Prp8 in active spliceosomes and together with suppressors of 5' and 3' splice site mutations this unambiguously locates the active site cavity. Structural and mechanistic similarities with group II self-splicing introns have encouraged the notion that the spliceosome is at heart a ribozyme, and recently the ligands for two catalytic magnesium ions were identified within U6 snRNA. They position catalytic divalent metal ions in the same way as Domain V of group II intron RNA, suggesting that the spliceosome and group II intron use the same catalytic mechanisms.


Subject(s)
Spliceosomes/chemistry , Spliceosomes/metabolism , Catalytic Domain , Evolution, Molecular , Humans , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...