Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6429, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37833274

ABSTRACT

RNA-binding proteins (RBPs) are crucial regulators of gene expression, often composed of defined domains interspersed with flexible, intrinsically disordered regions. Determining the structure of ribonucleoprotein (RNP) complexes involving such RBPs necessitates integrative structural modeling due to their lack of a single stable state. In this study, we integrate magnetic resonance, mass spectrometry, and small-angle scattering data to determine the solution structure of the polypyrimidine-tract binding protein 1 (PTBP1/hnRNP I) bound to an RNA fragment from the internal ribosome entry site (IRES) of the encephalomyocarditis virus (EMCV). This binding, essential for enhancing the translation of viral RNA, leads to a complex structure that demonstrates RNA and protein compaction, while maintaining pronounced conformational flexibility. Acting as an RNA chaperone, PTBP1 orchestrates the IRES RNA into a few distinct conformations, exposing the RNA stems outward. This conformational diversity is likely common among RNP structures and functionally important. Our approach enables atomic-level characterization of heterogeneous RNP structures.


Subject(s)
Internal Ribosome Entry Sites , RNA-Binding Proteins , RNA-Binding Proteins/metabolism , Encephalomyocarditis virus/genetics , RNA, Viral/metabolism , Nucleic Acid Conformation , Protein Biosynthesis
2.
Nucleic Acids Res ; 48(8): 4521-4537, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32170319

ABSTRACT

The polypyrimidine tract binding protein (PTB) is a multi-domain protein involved in alternative splicing, mRNA localization, stabilization, polyadenylation and translation initiation from internal ribosome entry sites (IRES). In this latter process, PTB promotes viral translation by interacting extensively with complex structured regions in the 5'-untranslated regions of viral RNAs at pyrimidine-rich targets located in single strand and hairpin regions. To better understand how PTB recognizes structured elements in RNA targets, we solved the solution structure of the N-terminal RNA recognition motif (RRM) in complex with an RNA hairpin embedding the loop sequence UCUUU, which is frequently found in IRESs of the picornovirus family. Surprisingly, a new three-turn α3 helix C-terminal to the RRM, folds upon binding the RNA hairpin. Although α3 does not mediate any contacts to the RNA, it acts as a sensor of RNA secondary structure, suggesting a role for RRM1 in detecting pyrimidine tracts in the context of structured RNA. Moreover, the degree of helix formation depends on the RNA loop sequence. Finally, we show that the α3 helix region, which is highly conserved in vertebrates, is crucial for PTB function in enhancing Encephalomyocarditis virus IRES activity.


Subject(s)
Polypyrimidine Tract-Binding Protein/chemistry , RNA Recognition Motif , RNA/chemistry , Encephalomyocarditis virus/genetics , Internal Ribosome Entry Sites , Models, Molecular , Mutation , Nucleic Acid Conformation , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Protein Binding , Protein Conformation, alpha-Helical , RNA/metabolism
3.
EMBO J ; 37(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29449323

ABSTRACT

The accurate cleavage of pre-micro(mi)RNAs by Dicer and mi/siRNA guide strand selection are important steps in forming the RNA-induced silencing complex (RISC). The role of Dicer binding partner TRBP in these processes remains poorly understood. Here, we solved the solution structure of the two N-terminal dsRNA binding domains (dsRBDs) of TRBP in complex with a functionally asymmetric siRNA using NMR, EPR, and single-molecule spectroscopy. We find that siRNA recognition by the dsRBDs is not sequence-specific but rather depends on the RNA shape. The two dsRBDs can swap their binding sites, giving rise to two equally populated, pseudo-symmetrical complexes, showing that TRBP is not a primary sensor of siRNA asymmetry. Using our structure to model a Dicer-TRBP-siRNA ternary complex, we show that TRBP's dsRBDs and Dicer's RNase III domains bind a canonical 19 base pair siRNA on opposite sides, supporting a mechanism whereby TRBP influences Dicer-mediated cleavage accuracy by binding the dsRNA region of the pre-miRNA during Dicer cleavage.


Subject(s)
Double-Stranded RNA Binding Motif , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Models, Molecular , Spectrum Analysis/methods
4.
RNA ; 21(11): 1931-42, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26370582

ABSTRACT

The human hnRNP C is a ubiquitous cellular protein involved in mRNA maturation. Recently, we have shown that this protein specifically recognizes uridine (U) pentamers through its single RNA recognition motif (RRM). However, a large fraction of natural RNA targets of hnRNP C consists of much longer contiguous uridine stretches. To understand how these extended sites are recognized, we studied the binding of the RRM to U-tracts of 8-11 bases. In vivo investigation of internal translation activation of unr (upstream of N-ras) mRNA indicates that the conservation of the entire hnRNP C binding site, UC(U)8, is required for hnRNP C-dependent IRES activation. The assays further suggest a synergistic interplay between hnRNP C monomers, dependent on the protein's ability to oligomerize. In vitro spectroscopic and thermodynamic analyses show that isolated RRMs bind to (U)11 oligomers as dimers. Structural modeling of a ternary double-RRM/RNA complex indicates additionally that two RRM copies can be accommodated on the canonical sequence UC(U)8. The proposed tandem RRM binding is in very good agreement with the transcriptome-wide recognition of extended U-tracts by full-length hnRNP C, which displays a cross-linking pattern consistent with a positively cooperative RRM dimer binding model.


Subject(s)
Binding Sites/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Protein Binding/genetics , RNA Recognition Motif Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Transcriptome/genetics , Uridine/genetics , Uridine/metabolism
5.
J Mol Biol ; 427(19): 3001-22, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26051023

ABSTRACT

The RNA recognition motif (RRM) is the far most abundant RNA binding domain. In addition to the typical ß1α1ß2ß3α2ß4 fold, various sub-structural elements have been described and reportedly contribute to the high functional versatility of RRMs. The heterogeneous nuclear ribonucleoprotein L (hnRNP L) is a highly abundant protein of 64 kDa comprising four RRM domains. Involved in many aspects of RNA metabolism, hnRNP L specifically binds to RNAs containing CA repeats or CA-rich clusters. However, a comprehensive structural description of hnRNP L including its sub-structural elements is missing. Here, we present the structural characterization of the RRM domains of hnRNP L and demonstrate their function in repressing exon 4 of SLC2A2. By comparison of the sub-structural elements between the two highly similar paralog families of hnRNP L and PTB, we defined signatures underlying interacting C-terminal coils (ICCs), the RRM34 domain interaction and RRMs with a C-terminal fifth ß-strand, a variation we denoted vRRMs. Furthermore, computational analysis revealed new putative ICC-containing RRM families and allowed us to propose an evolutionary scenario explaining the origins of the ICC and fifth ß-strand sub-structural extensions. Our studies provide insights of domain requirements in alternative splicing mediated by hnRNP L and molecular descriptions for the sub-structural elements. In addition, the analysis presented may help to classify other abundant RRM extensions and to predict structure-function relationships.


Subject(s)
Exons , Glucose Transporter Type 2/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/chemistry , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , RNA/metabolism , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA/genetics
6.
J Am Chem Soc ; 136(41): 14536-44, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25216038

ABSTRACT

HnRNP C is a ubiquitous RNA regulatory factor and the principal constituent of the nuclear hnRNP core particle. The protein contains one amino-terminal RNA recognition motif (RRM) known to bind uridine (U)-rich sequences. This work provides a molecular and mechanistic understanding of this interaction. We solved the solution structures of the RRM in complex with poly(U) oligomers of five and seven nucleotides. The five binding pockets of RRM recognize uridines with an unusual 5'-to-3' gradient of base selectivity. The target recognition is therefore strongly sensitive to base clustering, explaining the preference for contiguous uridine tracts. Using a novel approach integrating the structurally derived recognition consensus of the RRM with a thermodynamic description of its multi-register binding, we modeled the saturation of cellular uridine tracts by this protein. The binding pattern is remarkably consistent with the experimentally observed transcriptome-wide cross-link distribution of the full-length hnRNP C on short uridine tracts. This result re-establishes the RRM as the primary RNA-binding domain of the hnRNP C tetramer and provides a proof of concept for interpreting high-throughput interaction data using structural approaches.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group C/chemistry , Poly U/chemistry , RNA/chemistry , Humans , Kinetics , Models, Molecular , Molecular Structure , Thermodynamics
7.
Nucleic Acids Res ; 41(18): e172, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23921634

ABSTRACT

The three-dimensional structure determination of RNAs by NMR spectroscopy relies on chemical shift assignment, which still constitutes a bottleneck. In order to develop more efficient assignment strategies, we analysed relationships between sequence and (1)H and (13)C chemical shifts. Statistics of resonances from regularly Watson-Crick base-paired RNA revealed highly characteristic chemical shift clusters. We developed two approaches using these statistics for chemical shift assignment of double-stranded RNA (dsRNA): a manual approach that yields starting points for resonance assignment and simplifies decision trees and an automated approach based on the recently introduced automated resonance assignment algorithm FLYA. Both strategies require only unlabeled RNAs and three 2D spectra for assigning the H2/C2, H5/C5, H6/C6, H8/C8 and H1'/C1' chemical shifts. The manual approach proved to be efficient and robust when applied to the experimental data of RNAs with a size between 20 nt and 42 nt. The more advanced automated assignment approach was successfully applied to four stem-loop RNAs and a 42 nt siRNA, assigning 92-100% of the resonances from dsRNA regions correctly. This is the first automated approach for chemical shift assignment of non-exchangeable protons of RNA and their corresponding (13)C resonances, which provides an important step toward automated structure determination of RNAs.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , RNA, Double-Stranded/chemistry , Algorithms , Data Interpretation, Statistical , Databases, Nucleic Acid , Nucleic Acid Conformation , RNA, Small Interfering/chemistry , Software
8.
J Biomol NMR ; 52(2): 141-50, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22143941

ABSTRACT

A five-dimensional (5D) APSY (automated projection spectroscopy) HCNCH experiment is presented, which allows unambiguous correlation of sugar to base nuclei in nucleic acids. The pulse sequence uses multiple quantum (MQ) evolution which enables long constant-time evolution periods in all dimensions, an improvement that can also benefit non-APSY applications. Applied with an RNA with 23 nucleotides the 5D APSY-HCNCH experiment produced a complete and highly precise 5D chemical shift list within 1.5 h. Alternatively, and for molecules where the out-and-stay 5D experiment sensitivity is not sufficient, a set of out-and-back 3D APSY-HCN experiments is proposed: an intra-base (3D APSY-b-HCN) experiment in an MQ or in a TROSY version, and an MQ sugar-to-base (3D APSY-s-HCN) experiment. The two 3D peak lists require subsequent matching via the N1/9 chemical shift values to one 5D peak list. Optimization of the 3D APSY experiments for maximal precision in the N1/9 dimension allowed matching of all (15)N chemical shift values contained in both 3D peak lists. The precise 5D chemical shift correlation lists resulting from the 5D experiment or a pair of 3D experiments also provide a valuable basis for subsequent connection to chemical shifts derived with other experiments.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Nucleic Acid Conformation , RNA/chemistry , Inverted Repeat Sequences , Ribose/chemistry
9.
Mol Cell ; 41(5): 579-88, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21362553

ABSTRACT

Splicing of the c-src N1 exon is repressed by the polypyrimidine tract-binding protein (PTB or PTBP1). During exon repression, the U1 snRNP binds properly to the N1 exon 5' splice site but is made inactive by the presence of PTB. Examining the patterns of nuclease protection at this 5' splice site, we find that the interaction of U1 is altered by the adjacent PTB. Interestingly, UV crosslinking identifies a direct contact between the pre-mRNA-bound PTB and the U1 snRNA. EMSA, ITC, and NMR studies show that PTB RRMs 1 and 2 bind the pyrimidine-rich internal loop of U1 snRNA stem loop 4. The PTB/U1 interaction prevents further assembly of the U1 snRNP with spliceosomal components downstream. This precise interaction between a splicing regulator and an snRNA component of the spliceosome points to a range of different mechanisms for splicing regulation.


Subject(s)
Gene Expression Regulation , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing , RNA, Small Nuclear/genetics , Calorimetry/methods , Cross-Linking Reagents/chemistry , Exons , HeLa Cells , Humans , Magnetic Resonance Spectroscopy/methods , Models, Biological , Protein Structure, Tertiary , RNA, Messenger/metabolism , Ribonucleases/metabolism , Ultraviolet Rays
10.
Cell ; 143(2): 225-37, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20946981

ABSTRACT

Sequence-dependent recognition of dsDNA-binding proteins is well understood, yet sequence-specific recognition of dsRNA by proteins remains largely unknown, despite their importance in RNA maturation pathways. Adenosine deaminases that act on RNA (ADARs) recode genomic information by the site-selective deamination of adenosine. Here, we report the solution structure of the ADAR2 double-stranded RNA-binding motifs (dsRBMs) bound to a stem-loop pre-mRNA encoding the R/G editing site of GluR-2. The structure provides a molecular basis for how dsRBMs recognize the shape, and also more surprisingly, the sequence of the dsRNA. The unexpected direct readout of the RNA primary sequence by dsRBMs is achieved via the minor groove of the dsRNA and this recognition is critical for both editing and binding affinity at the R/G site of GluR-2. More generally, our findings suggest a solution to the sequence-specific paradox faced by many dsRBM-containing proteins that are involved in post-transcriptional regulation of gene expression.


Subject(s)
Adenosine Deaminase/chemistry , RNA, Double-Stranded/chemistry , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , RNA Precursors/metabolism , RNA, Double-Stranded/metabolism , RNA-Binding Proteins , Rats , Receptors, AMPA/genetics , Sequence Alignment
11.
Nucleic Acids Res ; 38(20): e188, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20798173

ABSTRACT

Structural information on RNA, emerging more and more as a major regulator in gene expression, dramatically lags behind compared with information on proteins. Although NMR spectroscopy has proven to be an excellent tool to solve RNA structures, it is hampered by the severe spectral resonances overlap found in RNA, limiting its use for large RNA molecules. Segmental isotope labeling of RNA or ligation of a chemically synthesized RNA containing modified nucleotides with an unmodified RNA fragment have proven to have high potential in overcoming current limitations in obtaining structural information on RNA. However, low yields, cumbersome preparations and sequence requirements have limited its broader application in structural biology. Here we present a fast and efficient approach to generate multiple segmentally labeled RNAs with virtually no sequence requirements with very high yields (up to 10-fold higher than previously reported). We expect this approach to open new avenues in structural biology of RNA.


Subject(s)
Isotope Labeling/methods , RNA, Catalytic , RNA/chemistry , Ribonuclease H , Chromatography, High Pressure Liquid , DNA Ligases , Nuclear Magnetic Resonance, Biomolecular , RNA/metabolism , RNA, Untranslated/chemistry , Transcription, Genetic
12.
EMBO J ; 25(11): 2487-97, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16710302

ABSTRACT

Regulatory RNA elements, like riboswitches, respond to intracellular signals by three-dimensional (3D) conformational changes. RNA thermometers employ a similar strategy to sense temperature changes in the cell and regulate the translational machinery. We present here the first 3D NMR structure of the functional domain of a highly conserved bacterial RNA thermometer containing the ribosome binding site that remains occluded at normal temperatures (30 degrees C). We identified a region adjacent to the Shine-Dalgarno sequence that has a network of weak hydrogen bonds within the RNA helix. With the onset of heat shock at 42 degrees C, destabilisation of the RNA structure initiates at this region and favours the release of the ribosome binding site and of the start codon. Deletion of a highly conserved G residue leads to the formation of a stable regular RNA helix that loses thermosensing ability. Our results indicate that RNA thermometers are able to sense temperature changes without the aid of accessory factors.


Subject(s)
Nucleic Acid Conformation , RNA/chemistry , Temperature , Thermometers , Base Sequence , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , RNA/metabolism
13.
FEBS J ; 272(9): 2118-31, 2005 May.
Article in English | MEDLINE | ID: mdl-15853797

ABSTRACT

The RNA recognition motif (RRM), also known as RNA-binding domain (RBD) or ribonucleoprotein domain (RNP) is one of the most abundant protein domains in eukaryotes. Based on the comparison of more than 40 structures including 15 complexes (RRM-RNA or RRM-protein), we reviewed the structure-function relationships of this domain. We identified and classified the different structural elements of the RRM that are important for binding a multitude of RNA sequences and proteins. Common structural aspects were extracted that allowed us to define a structural leitmotif of the RRM-nucleic acid interface with its variations. Outside of the two conserved RNP motifs that lie in the center of the RRM beta-sheet, the two external beta-strands, the loops, the C- and N-termini, or even a second RRM domain allow high RNA-binding affinity and specific recognition. Protein-RRM interactions that have been found in several structures reinforce the notion of an extreme structural versatility of this domain supporting the numerous biological functions of the RRM-containing proteins.


Subject(s)
Amino Acid Sequence , Gene Expression Regulation , Protein Biosynthesis , Protein Conformation , Protein Folding , Models, Molecular , Molecular Sequence Data , Protein Binding , RNA/metabolism , Sequence Alignment
14.
RNA ; 11(2): 173-86, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659357

ABSTRACT

We have solved the NMR structure of the 31-nucleotide (nt) apoB mRNA stem-loop, a substrate of the cytidine deaminase APOBEC1. We found that the edited base located at the 5' end of the octa-loop is stacked between two adenosines in both the unedited (cytidine 6666) and the edited (uridine 6666) forms and that the rest of the loop is unstructured. The 11-nt "mooring" sequence essential for editing is partially flexible although it is mostly in the stem of the RNA. The octa-loop and the internal loop in the middle of the stem confer this flexibility. These findings shed light on why APOBEC1 alone cannot edit efficiently the cytidine 6666 under physiological conditions, the editing base being buried in the loop and not directly accessible. We also show that APOBEC1 does not specifically bind apoB mRNA and requires the auxiliary factor, APOBEC1 complementary factor (ACF), to edit specifically cytidine 6666. The binding of ACF to both the mooring sequence and APOBEC1 explains the specificity of the reaction. Our NMR study lead us to propose a mechanism in which ACF recognizes first the flexible nucleotides of the mooring sequence (the internal loop and the 3' end octa-loop) and subsequently melts the stem-loop, exposing the amino group of the cytidine 6666 to APOBEC1. Thus, the flexibility of the mooring sequence plays a central role in the RNA recognition by ACF.


Subject(s)
Apolipoproteins B/genetics , Cytidine Deaminase/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , APOBEC-1 Deaminase , Base Sequence , Humans , In Vitro Techniques , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , RNA Editing , RNA, Messenger/genetics , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...