Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Cell ; 83(13): 2258-2275.e11, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37369199

ABSTRACT

The pre-mRNA life cycle requires intron processing; yet, how intron-processing defects influence splicing and gene expression is unclear. Here, we find that TTDN1/MPLKIP, which is encoded by a gene implicated in non-photosensitive trichothiodystrophy (NP-TTD), functionally links intron lariat processing to spliceosomal function. The conserved TTDN1 C-terminal region directly binds lariat debranching enzyme DBR1, whereas its N-terminal intrinsically disordered region (IDR) binds the intron-binding complex (IBC). TTDN1 loss, or a mutated IDR, causes significant intron lariat accumulation, as well as splicing and gene expression defects, mirroring phenotypes observed in NP-TTD patient cells. A Ttdn1-deficient mouse model recapitulates intron-processing defects and certain neurodevelopmental phenotypes seen in NP-TTD. Fusing DBR1 to the TTDN1 IDR is sufficient to recruit DBR1 to the IBC and circumvents the functional requirement for TTDN1. Collectively, our findings link RNA lariat processing with splicing outcomes by revealing the molecular function of TTDN1.


Subject(s)
Trichothiodystrophy Syndromes , Animals , Mice , Introns/genetics , Trichothiodystrophy Syndromes/genetics , RNA Nucleotidyltransferases/genetics , RNA Splicing
2.
Nature ; 617(7962): 842-850, 2023 May.
Article in English | MEDLINE | ID: mdl-37165190

ABSTRACT

Pre-mRNA splicing follows a pathway driven by ATP-dependent RNA helicases. A crucial event of the splicing pathway is the catalytic activation, which takes place at the transition between the activated Bact and the branching-competent B* spliceosomes. Catalytic activation occurs through an ATP-dependent remodelling mediated by the helicase PRP2 (also known as DHX16)1-3. However, because PRP2 is observed only at the periphery of spliceosomes3-5, its function has remained elusive. Here we show that catalytic activation occurs in two ATP-dependent stages driven by two helicases: PRP2 and Aquarius. The role of Aquarius in splicing has been enigmatic6,7. Here the inactivation of Aquarius leads to the stalling of a spliceosome intermediate-the BAQR complex-found halfway through the catalytic activation process. The cryogenic electron microscopy structure of BAQR reveals how PRP2 and Aquarius remodel Bact and BAQR, respectively. Notably, PRP2 translocates along the intron while it strips away the RES complex, opens the SF3B1 clamp and unfastens the branch helix. Translocation terminates six nucleotides downstream of the branch site through an assembly of PPIL4, SKIP and the amino-terminal domain of PRP2. Finally, Aquarius enables the dissociation of PRP2, plus the SF3A and SF3B complexes, which promotes the relocation of the branch duplex for catalysis. This work elucidates catalytic activation in human splicing, reveals how a DEAH helicase operates and provides a paradigm for how helicases can coordinate their activities.


Subject(s)
Biocatalysis , RNA Splicing , Humans , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Cyclophilins/metabolism , RNA Precursors/metabolism , RNA Splicing Factors/metabolism , RNA-Binding Proteins/metabolism , Spliceosomes/metabolism
3.
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
4.
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
5.
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
6.
Nature ; 529(7585): 231-4, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26735012

ABSTRACT

Catalysis in biology is restricted to RNA (ribozymes) and protein enzymes, but synthetic biomolecular catalysts can also be made of DNA (deoxyribozymes) or synthetic genetic polymers. In vitro selection from synthetic random DNA libraries identified DNA catalysts for various chemical reactions beyond RNA backbone cleavage. DNA-catalysed reactions include RNA and DNA ligation in various topologies, hydrolytic cleavage and photorepair of DNA, as well as reactions of peptides and small molecules. In spite of comprehensive biochemical studies of DNA catalysts for two decades, fundamental mechanistic understanding of their function is lacking in the absence of three-dimensional models at atomic resolution. Early attempts to solve the crystal structure of an RNA-cleaving deoxyribozyme resulted in a catalytically irrelevant nucleic acid fold. Here we report the crystal structure of the RNA-ligating deoxyribozyme 9DB1 (ref. 14) at 2.8 Å resolution. The structure captures the ligation reaction in the post-catalytic state, revealing a compact folding unit stabilized by numerous tertiary interactions, and an unanticipated organization of the catalytic centre. Structure-guided mutagenesis provided insights into the basis for regioselectivity of the ligation reaction and allowed remarkable manipulation of substrate recognition and reaction rate. Moreover, the structure highlights how the specific properties of deoxyribose are reflected in the backbone conformation of the DNA catalyst, in support of its intricate three-dimensional organization. The structural principles underlying the catalytic ability of DNA elucidate differences and similarities in DNA versus RNA catalysts, which is relevant for comprehending the privileged position of folded RNA in the prebiotic world and in current organisms.


Subject(s)
DNA, Catalytic/chemistry , Nucleic Acid Conformation , Base Sequence , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , DNA, Catalytic/chemical synthesis , DNA, Catalytic/metabolism , Deoxyribose/chemistry , Deoxyribose/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Nucleotides/chemistry , Nucleotides/metabolism , Polynucleotide Ligases/chemistry , Polynucleotide Ligases/metabolism , RNA/chemistry , RNA/metabolism , RNA Folding , Substrate Specificity
7.
Mol Cell ; 35(4): 454-66, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19716790

ABSTRACT

Brr2 is a unique DExD/H box protein required for catalytic activation and disassembly of the spliceosome. It contains two tandem helicase cassettes that both comprise dual RecA-like domains and a noncanonical Sec63 unit. The latter may bestow the enzyme with unique properties. We have determined crystal structures of the C-terminal Sec63 unit of yeast Brr2, revealing three domains, two of which resemble functional modules of a DNA helicase, Hel308, despite lacking significant sequence similarity. This structural similarity together with sequence conservation between the enzymes throughout the RecA-like domains and a winged helix domain allowed us to devise a structural model of the N-terminal active cassette of Brr2. We consolidated the model by rational mutagenesis combined with splicing and U4/U6 di-snRNA unwinding assays, highlighting how the RecA-like domains and the Sec63 unit form a functional entity that appears suitable for unidirectional and processive RNA duplex unwinding during spliceosome activation and disassembly.


Subject(s)
DNA Helicases/chemistry , Fungal Proteins/chemistry , RNA Helicases/chemistry , Spliceosomes/enzymology , Yeasts/enzymology , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA Helicases/genetics , DNA Helicases/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Structure, Tertiary , RNA/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , Structure-Activity Relationship , Time Factors , Yeasts/genetics
8.
EMBO J ; 31(9): 2222-34, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22407296

ABSTRACT

The yeast splicing factor Cwc2 contacts several catalytically important RNA elements in the active spliceosome, suggesting that Cwc2 is involved in determining their spatial arrangement at the spliceosome's catalytic centre. We have determined the crystal structure of the Cwc2 functional core, revealing how a previously uncharacterized Torus domain, an RNA recognition motif (RRM) and a zinc finger (ZnF) are tightly integrated in a compact folding unit. The ZnF plays a pivotal role in the architecture of the whole assembly. UV-induced crosslinking of Cwc2-U6 snRNA allowed the identification by mass spectrometry of six RNA-contacting sites: four in or close to the RRM domain, one in the ZnF and one on a protruding element connecting the Torus and RRM domains. The three distinct regions contacting RNA are connected by a contiguous and conserved positively charged surface, suggesting an expanded interface for RNA accommodation. Cwc2 mutations confirmed that the connector element plays a crucial role in splicing. We conclude that Cwc2 acts as a multipartite RNA-binding platform to bring RNA elements of the spliceosome's catalytic centre into an active conformation.


Subject(s)
RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Base Sequence , Molecular Sequence Data , Nucleotide Motifs , Protein Folding , Protein Structure, Tertiary , RNA Precursors/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Zinc Fingers
9.
Proc Natl Acad Sci U S A ; 109(43): 17418-23, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23045696

ABSTRACT

Assembly of a spliceosome, catalyzing precursor-messenger RNA splicing, involves multiple RNA-protein remodeling steps, driven by eight conserved DEXD/H-box RNA helicases. The 250-kDa Brr2 enzyme, which is essential for U4/U6 di-small nuclear ribonucleoprotein disruption during spliceosome catalytic activation and for spliceosome disassembly, is the only member of this group that is permanently associated with the spliceosome, thus requiring its faithful regulation. At the same time, Brr2 represents a unique subclass of superfamily 2 nucleic acid helicases, containing tandem helicase cassettes. Presently, the mechanistic and regulatory consequences of this unconventional architecture are unknown. Here we show that in human Brr2, two ring-like helicase cassettes intimately interact and functionally cooperate and how retinitis pigmentosa-linked Brr2 mutations interfere with the enzyme's function. Only the N-terminal cassette harbors ATPase and helicase activities in isolation. Comparison with other helicases and mutational analyses show how it threads single-stranded RNA, and structural features suggest how it can load onto an internal region of U4/U6 di-snRNA. Although the C-terminal cassette does not seem to engage RNA in the same fashion, it binds ATP and strongly stimulates the N-terminal helicase. Mutations at the cassette interface, in an intercassette linker or in the C-terminal ATP pocket, affect this cross-talk in diverse ways. Together, our results reveal the structural and functional interplay between two helicase cassettes in a tandem superfamily 2 enzyme and point to several sites through which Brr2 activity may be regulated.


Subject(s)
Ribonucleoproteins, Small Nuclear/chemistry , Spliceosomes , Catalysis , DNA Helicases/metabolism , Humans , Models, Molecular , Mutation , Protein Conformation , Retinitis Pigmentosa/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism
10.
EMBO J ; 27(21): 2929-40, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18843295

ABSTRACT

Precursor-messenger RNA (pre-mRNA) splicing encompasses two sequential transesterification reactions in distinct active sites of the spliceosome that are transiently established by the interplay of small nuclear (sn) RNAs and spliceosomal proteins. Protein Prp8 is an active site component but the molecular mechanisms, by which it might facilitate splicing catalysis, are unknown. We have determined crystal structures of corresponding portions of yeast and human Prp8 that interact with functional regions of the pre-mRNA, revealing a phylogenetically conserved RNase H fold, augmented by Prp8-specific elements. Comparisons to RNase H-substrate complexes suggested how an RNA encompassing a 5'-splice site (SS) could bind relative to Prp8 residues, which on mutation, suppress splice defects in pre-mRNAs and snRNAs. A truncated RNase H-like active centre lies next to a known contact region of the 5'SS and directed mutagenesis confirmed that this centre is a functional hotspot. These data suggest that Prp8 employs an RNase H domain to help assemble and stabilize the spliceosomal catalytic core, coordinate the activities of other splicing factors and possibly participate in chemical catalysis of splicing.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Ribonuclease H/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Conserved Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptides/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Pyrimidines , RNA Precursors/metabolism , RNA Splice Sites , RNA-Binding Proteins , Ribonucleoprotein, U4-U6 Small Nuclear , Ribonucleoprotein, U5 Small Nuclear , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Structure-Activity Relationship
11.
RNA Biol ; 9(11): 1311-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064115

ABSTRACT

Pre-mRNA splicing occurs in two chemical steps that are catalyzed by a large, dynamic RNA-protein complex called the spliceosome. Initially assembled in a catalytically inactive form, the spliceosome undergoes massive compositional and conformational remodeling, through which disparate RNA elements are re-configured and juxtaposed into a functional catalytic center. The intricate construction of the catalytic center requires the assistance of spliceosomal proteins. Recent structure-function analyses have demonstrated that the yeast-splicing factor Cwc2 is a main player that contacts and shapes the catalytic center of the spliceosome into a functional conformation. With this advance, corroborated by the atomic structure of the evolutionarily related group IIC introns, our understanding of the organization and formation of the spliceosomal catalytic center has progressed to a new level.


Subject(s)
RNA Precursors/genetics , RNA Splice Sites , RNA Splicing/genetics , RNA, Messenger/genetics , Spliceosomes/genetics , Carrier Proteins/genetics , Catalysis , Catalytic Domain/genetics , Humans , Introns/genetics , Nucleic Acid Conformation , RNA, Small Nuclear/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure
12.
Sci Adv ; 8(28): eabm9875, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35857496

ABSTRACT

RNA polymerase III (RNAPIII) synthesizes essential and abundant noncoding RNAs such as transfer RNAs. Controlling RNAPIII span of activity by accurate and efficient termination is a challenging necessity to ensure robust gene expression and to prevent conflicts with other DNA-associated machineries. The mechanism of RNAPIII termination is believed to be simpler than that of other eukaryotic RNA polymerases, solely relying on the recognition of a T-tract in the nontemplate strand. Here, we combine high-resolution genome-wide analyses and in vitro transcription termination assays to revisit the mechanism of RNAPIII transcription termination in budding yeast. We show that T-tracts are necessary but not always sufficient for termination and that secondary structures of the nascent RNAs are important auxiliary cis-acting elements. Moreover, we show that the helicase Sen1 plays a key role in a fail-safe termination pathway. Our results provide a comprehensive model illustrating how multiple mechanisms cooperate to ensure efficient RNAPIII transcription termination.


Subject(s)
RNA Polymerase III , Saccharomyces cerevisiae Proteins , DNA Helicases/metabolism , Genome-Wide Association Study , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
13.
Nat Commun ; 12(1): 3549, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112799

ABSTRACT

Fluorogenic RNA aptamers are synthetic functional RNAs that specifically bind and activate conditional fluorophores. The Chili RNA aptamer mimics large Stokes shift fluorescent proteins and exhibits high affinity for 3,5-dimethoxy-4-hydroxybenzylidene imidazolone (DMHBI) derivatives to elicit green or red fluorescence emission. Here, we elucidate the structural and mechanistic basis of fluorescence activation by crystallography and time-resolved optical spectroscopy. Two co-crystal structures of the Chili RNA with positively charged DMHBO+ and DMHBI+ ligands revealed a G-quadruplex and a trans-sugar-sugar edge G:G base pair that immobilize the ligand by π-π stacking. A Watson-Crick G:C base pair in the fluorophore binding site establishes a short hydrogen bond between the N7 of guanine and the phenolic OH of the ligand. Ultrafast excited state proton transfer (ESPT) from the neutral chromophore to the RNA was found with a time constant of 130 fs and revealed the mode of action of the large Stokes shift fluorogenic RNA aptamer.


Subject(s)
Aptamers, Nucleotide/chemistry , G-Quadruplexes , Guanine/chemistry , RNA/chemistry , Binding Sites , Crystallography , Fluorescence , Hydrogen Bonding , Ligands , Magnetic Resonance Spectroscopy , Molecular Structure , Mutagenesis , Mutation , Protons , Spectrometry, Fluorescence
14.
Nat Commun ; 12(1): 4491, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34301950

ABSTRACT

Intron selection during the formation of prespliceosomes is a critical event in pre-mRNA splicing. Chemical modulation of intron selection has emerged as a route for cancer therapy. Splicing modulators alter the splicing patterns in cells by binding to the U2 snRNP (small nuclear ribonucleoprotein)-a complex chaperoning the selection of branch and 3' splice sites. Here we report crystal structures of the SF3B module of the U2 snRNP in complex with spliceostatin and sudemycin FR901464 analogs, and the cryo-electron microscopy structure of a cross-exon prespliceosome-like complex arrested with spliceostatin A. The structures reveal how modulators inactivate the branch site in a sequence-dependent manner and stall an E-to-A prespliceosome intermediate by covalent coupling to a nucleophilic zinc finger belonging to the SF3B subunit PHF5A. These findings support a mechanism of intron recognition by the U2 snRNP as a toehold-mediated strand invasion and advance an unanticipated drug targeting concept.


Subject(s)
DNA/genetics , Introns/genetics , Pyrans/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spiro Compounds/metabolism , Spliceosomes/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Humans , Lactones/chemistry , Lactones/metabolism , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Pyrans/chemistry , Pyrones/chemistry , Pyrones/metabolism , Ribonucleoprotein, U2 Small Nuclear/chemistry , Spiro Compounds/chemistry , Spliceosomes/ultrastructure
15.
Sci Rep ; 10(1): 9828, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32555289

ABSTRACT

Cohesin is a protein complex whose core subunits, Smc1, Smc3, Scc1, and SA1/SA2 form a ring-like structure encircling the DNA. Cohesins play a key role in the expression, repair, and segregation of eukaryotic genomes. Following a catalytic mechanism that is insufficiently understood, Esco1 and Esco2 acetyltransferases acetylate the cohesin subunit Smc3, thereby inducing stabilization of cohesin on DNA. As a prerequisite for structure-guided investigation of enzymatic activity, we determine here the crystal structure of the mouse Esco2/CoA complex at 1.8 Šresolution. We reconstitute cohesin as tri- or tetrameric assemblies and use those as physiologically-relevant substrates for enzymatic assays in vitro. Furthermore, we employ cell-based complementation studies in mouse embryonic fibroblast deficient for Esco1 and Esco2, as a means to identify catalytically-important residues in vivo. These analyses demonstrate that D567/S566 and E491/S527, located on opposite sides of the murine Esco2 active site cleft, are critical for catalysis. Our experiments support a catalytic mechanism of acetylation where residues D567 and E491 are general bases that deprotonate the ε-amino group of lysine substrate, also involving two nearby serine residues - S566 and S527- that possess a proton relay function.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Biocatalysis , Catalytic Domain , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Acetylation , Acetyltransferases/genetics , Amino Acid Sequence , Animals , Chromosomal Proteins, Non-Histone/genetics , Coenzyme A/metabolism , Humans , Mice , Models, Molecular , Mutation
16.
Protein Expr Purif ; 65(1): 30-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19111619

ABSTRACT

The protein giant neurofibromin (320kDa) is the protein product of the NF1 tumor suppressor gene, alterations of which are responsible for the pathogenesis of neurofibromatosis type 1 (NF1). Neurofibromin is a Ras-specific GTPase activating protein (RasGAP) that, 15 years after the cloning of the gene, remains the only clearly defined function of the protein. In a structural proteomics approach, we aimed at defining functions beyond RasGAP activity based on the discovery of structural modules. Given the poor outcome of domain prediction tools, we have undertaken a fragment solubility survey covering the full protein sequence, with the aim of defining new domain boundaries or fragments that could be investigated by biochemical methods including structural analysis. More than 200 constructs have been expressed and tested for solubility in small scale assays. Boundaries were chosen based upon secondary structure predictions, sequence conservation among neurofibromin orthologues and chemical properties of amino acids. Using this strategy we recently discovered a novel bipartite module in neurofibromin. We have expanded our study to include ESPRIT, a library-based construct screen, to perform fragment testing at a finer level with respect to the choice of terminal residues. Our study confirms earlier notions about the challenges neurofibromin presents to the biochemist and points to strategies whereby the success rate may be enhanced in the future.


Subject(s)
Neurofibromatosis 1/enzymology , Neurofibromin 1/chemistry , Humans , Neurofibromin 1/biosynthesis , Neurofibromin 1/genetics , Neurofibromin 1/isolation & purification , Protein Stability , Protein Structure, Tertiary/physiology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Solubility
17.
Wiley Interdiscip Rev RNA ; 7(2): 259-74, 2016.
Article in English | MEDLINE | ID: mdl-26874649

ABSTRACT

Splicing is an essential step of gene expression. It occurs in two consecutive chemical reactions catalyzed by a large protein-RNA complex named the spliceosome. Assembled on the pre-mRNA substrate from five small nuclear proteins, the spliceosome acts as a protein-controlled ribozyme to catalyze the two reactions and finally dissociates into its components, which are re-used for a new round of splicing. Upon following this cyclic pathway, the spliceosome undergoes numerous intermediate stages that differ in composition as well as in their internal RNA-RNA and RNA-protein contacts. The driving forces and control mechanisms of these remodeling processes are provided by specific molecular motors called RNA helicases. While eight spliceosomal helicases are present in all organisms, higher eukaryotes contain five additional ones potentially required to drive a more intricate splicing pathway and link it to an RNA metabolism of increasing complexity. Spliceosomal helicases exhibit a notable structural diversity in their accessory domains and overall architecture, in accordance with the diversity of their task-specific functions. This review summarizes structure-function knowledge about all spliceosomal helicases, including the latter five, which traditionally are treated separately from the conserved ones. The implications of the structural characteristics of helicases for their functions, as well as for their structural communication within the multi-subunits environment of the spliceosome, are pointed out.


Subject(s)
RNA Helicases/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , Animals , DEAD-box RNA Helicases/metabolism , Humans , Protein Binding , RNA Helicases/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Spliceosomes/metabolism , Substrate Specificity
18.
Science ; 353(6306): 1399-1405, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27562955

ABSTRACT

The activated spliceosome (Bact) is in a catalytically inactive state and is remodeled into a catalytically active machine by the RNA helicase Prp2, but the mechanism is unclear. Here, we describe a 3D electron cryomicroscopy structure of the Saccharomyces cerevisiae Bact complex at 5.8-angstrom resolution. Our model reveals that in Bact, the catalytic U2/U6 RNA-Prp8 ribonucleoprotein core is already established, and the 5' splice site (ss) is oriented for step 1 catalysis but occluded by protein. The first-step nucleophile-the branchsite adenosine-is sequestered within the Hsh155 HEAT domain and is held 50 angstroms away from the 5'ss. Our structure suggests that Prp2 adenosine triphosphatase-mediated remodeling leads to conformational changes in Hsh155's HEAT domain that liberate the first-step reactants for catalysis.


Subject(s)
RNA, Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/ultrastructure , Spliceosomes/ultrastructure , Adenosine Triphosphatases , Biocatalysis , Catalytic Domain , Cryoelectron Microscopy , Exons , Protein Conformation , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Splice Sites , RNA Splicing , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/chemistry
19.
Nat Struct Mol Biol ; 22(2): 138-44, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25599396

ABSTRACT

Aquarius is a multifunctional putative RNA helicase that binds precursor-mRNA introns at a defined position. Here we report the crystal structure of human Aquarius, revealing a central RNA helicase core and several unique accessory domains, including an ARM-repeat domain. We show that Aquarius is integrated into spliceosomes as part of a pentameric intron-binding complex (IBC) that, together with the ARM domain, cross-links to U2 snRNP proteins within activated spliceosomes; this suggests that the latter aid in positioning Aquarius on the intron. Aquarius's ARM domain is essential for IBC formation, thus indicating that it has a key protein-protein-scaffolding role. Finally, we provide evidence that Aquarius is required for efficient precursor-mRNA splicing in vitro. Our findings highlight the remarkable structural adaptations of a helicase to achieve position-specific recruitment to a ribonucleoprotein complex and reveal a new building block of the human spliceosome.


Subject(s)
RNA Helicases/chemistry , RNA Helicases/metabolism , Spliceosomes/metabolism , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Crystallography, X-Ray , Humans , Introns/genetics , Protein Binding/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , RNA Helicases/genetics , RNA Splicing/genetics , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism
20.
Sci Rep ; 5: 17338, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626428

ABSTRACT

Full-length Aß1-42 and Aß1-40, N-truncated pyroglutamate Aß3-42 and Aß4-42 are major variants in the Alzheimer brain. Aß4-42 has not been considered as a therapeutic target yet. We demonstrate that the antibody NT4X and its Fab fragment reacting with both the free N-terminus of Aß4-x and pyroglutamate Aß3-X mitigated neuron loss in Tg4-42 mice expressing Aß4-42 and completely rescued spatial reference memory deficits after passive immunization. NT4X and its Fab fragment also rescued working memory deficits in wild type mice induced by intraventricular injection of Aß4-42. NT4X reduced pyroglutamate Aß3-x, Aßx-40 and Thioflavin-S positive plaque load after passive immunization of 5XFAD mice. Aß1-x and Aßx-42 plaque deposits were unchanged. Importantly, for the first time, we demonstrate that passive immunization using the antibody NT4X is therapeutically beneficial in Alzheimer mouse models showing that N-truncated Aß starting with position four in addition to pyroglutamate Aß3-x is a relevant target to fight Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunization, Passive/methods , Peptide Fragments/immunology , Alzheimer Disease/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Disease Models, Animal , Humans , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL