Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
PLoS Comput Biol ; 19(7): e1010774, 2023 07.
Article in English | MEDLINE | ID: mdl-37406007

ABSTRACT

Typical drug discovery and development processes are costly, time consuming and often biased by expert opinion. Aptamers are short, single-stranded oligonucleotides (RNA/DNA) that bind to target proteins and other types of biomolecules. Compared with small-molecule drugs, aptamers can bind to their targets with high affinity (binding strength) and specificity (uniquely interacting with the target only). The conventional development process for aptamers utilizes a manual process known as Systematic Evolution of Ligands by Exponential Enrichment (SELEX), which is costly, slow, dependent on library choice and often produces aptamers that are not optimized. To address these challenges, in this research, we create an intelligent approach, named DAPTEV, for generating and evolving aptamer sequences to support aptamer-based drug discovery and development. Using the COVID-19 spike protein as a target, our computational results suggest that DAPTEV is able to produce structurally complex aptamers with strong binding affinities.


Subject(s)
Aptamers, Nucleotide , COVID-19 , Humans , Aptamers, Nucleotide/chemistry , SELEX Aptamer Technique/methods , Drug Design , RNA , Ligands
2.
Methods Mol Biol ; 2568: 193-211, 2023.
Article in English | MEDLINE | ID: mdl-36227570

ABSTRACT

RNA three-dimensional structures provide rich and vital information for understanding their functions. Recent advances in cryogenic electron microscopy (cryo-EM) allow structure determination of RNAs and ribonucleoprotein (RNP) complexes. However, limited global and local resolutions of RNA cryo-EM maps pose great challenges in tracing RNA coordinates. The Rosetta-based "auto-DRRAFTER" method builds RNA models into moderate-resolution RNA cryo-EM density as part of the Ribosolve pipeline. Here, we describe a step-by-step protocol for auto-DRRAFTER using a glycine riboswitch from Fusobacterium nucleatum as an example. Successful implementation of this protocol allows automated RNA modeling into RNA cryo-EM density, accelerating our understanding of RNA structure-function relationships. Input and output files are being made available at https://github.com/auto-DRRAFTER/springer-chapter .


Subject(s)
RNA , Riboswitch , Cryoelectron Microscopy/methods , Glycine , Models, Molecular , Protein Conformation , Ribonucleoproteins
3.
Nature ; 610(7932): 575-581, 2022 10.
Article in English | MEDLINE | ID: mdl-36224386

ABSTRACT

RNA-guided systems, such as CRISPR-Cas, combine programmable substrate recognition with enzymatic function, a combination that has been used advantageously to develop powerful molecular technologies1,2. Structural studies of these systems have illuminated how the RNA and protein jointly recognize and cleave their substrates, guiding rational engineering for further technology development3. Recent work identified a new class of RNA-guided systems, termed OMEGA, which include IscB, the likely ancestor of Cas9, and the nickase IsrB, a homologue of IscB lacking the HNH nuclease domain4. IsrB consists of only around 350 amino acids, but its small size is counterbalanced by a relatively large RNA guide (roughly 300-nt ωRNA). Here, we report the cryogenic-electron microscopy structure of Desulfovirgula thermocuniculi IsrB (DtIsrB) in complex with its cognate ωRNA and a target DNA. We find the overall structure of the IsrB protein shares a common scaffold with Cas9. In contrast to Cas9, however, which uses a recognition (REC) lobe to facilitate target selection, IsrB relies on its ωRNA, part of which forms an intricate ternary structure positioned analogously to REC. Structural analyses of IsrB and its ωRNA as well as comparisons to other RNA-guided systems highlight the functional interplay between protein and RNA, advancing our understanding of the biology and evolution of these diverse systems.


Subject(s)
DNA , Deoxyribonuclease I , RNA, Guide, Kinetoplastida , CRISPR-Cas Systems , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/metabolism , Deoxyribonuclease I/ultrastructure , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism , RNA, Guide, Kinetoplastida/ultrastructure , Cryoelectron Microscopy , CRISPR-Associated Proteins/chemistry
4.
Proc Natl Acad Sci U S A ; 119(37): e2209146119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36067294

ABSTRACT

The Tetrahymena group I intron has been a key system in the understanding of RNA folding and misfolding. The molecule folds into a long-lived misfolded intermediate (M) in vitro, which has been known to form extensive native-like secondary and tertiary structures but is separated by an unknown kinetic barrier from the native state (N). Here, we used cryogenic electron microscopy (cryo-EM) to resolve misfolded structures of the Tetrahymena L-21 ScaI ribozyme. Maps of three M substates (M1, M2, M3) and one N state were achieved from a single specimen with overall resolutions of 3.5 Å, 3.8 Å, 4.0 Å, and 3.0 Å, respectively. Comparisons of the structures reveal that all the M substates are highly similar to N, except for rotation of a core helix P7 that harbors the ribozyme's guanosine binding site and the crossing of the strands J7/3 and J8/7 that connect P7 to the other elements in the ribozyme core. This topological difference between the M substates and N state explains the failure of 5'-splice site substrate docking in M, supports a topological isomer model for the slow refolding of M to N due to a trapped strand crossing, and suggests pathways for M-to-N refolding.


Subject(s)
RNA Folding , RNA, Catalytic , Tetrahymena , Cryoelectron Microscopy , Kinetics , RNA, Catalytic/chemistry , Tetrahymena/genetics
6.
Nature ; 596(7873): 603-607, 2021 08.
Article in English | MEDLINE | ID: mdl-34381213

ABSTRACT

Single-particle cryogenic electron microscopy (cryo-EM) has become a standard technique for determining protein structures at atomic resolution1-3. However, cryo-EM studies of protein-free RNA are in their early days. The Tetrahymena thermophila group I self-splicing intron was the first ribozyme to be discovered and has been a prominent model system for the study of RNA catalysis and structure-function relationships4, but its full structure remains unknown. Here we report cryo-EM structures of the full-length Tetrahymena ribozyme in substrate-free and bound states at a resolution of 3.1 Å. Newly resolved peripheral regions form two coaxially stacked helices; these are interconnected by two kissing loop pseudoknots that wrap around the catalytic core and include two previously unforeseen (to our knowledge) tertiary interactions. The global architecture is nearly identical in both states; only the internal guide sequence and guanosine binding site undergo a large conformational change and a localized shift, respectively, upon binding of RNA substrates. These results provide a long-sought structural view of a paradigmatic RNA enzyme and signal a new era for the cryo-EM-based study of structure-function relationships in ribozymes.


Subject(s)
Cryoelectron Microscopy , Nucleic Acid Conformation , RNA, Catalytic/chemistry , RNA, Catalytic/ultrastructure , Tetrahymena thermophila , Apoenzymes/chemistry , Apoenzymes/ultrastructure , Holoenzymes/chemistry , Holoenzymes/ultrastructure , Models, Molecular , Tetrahymena thermophila/enzymology , Tetrahymena thermophila/genetics
7.
Nat Commun ; 12(1): 2165, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846332

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing catalyzed by ADAR enzymes occurs in double-stranded RNAs. Despite a compelling need towards predictive understanding of natural and engineered editing events, how the RNA sequence and structure determine the editing efficiency and specificity (i.e., cis-regulation) is poorly understood. We apply a CRISPR/Cas9-mediated saturation mutagenesis approach to generate libraries of mutations near three natural editing substrates at their endogenous genomic loci. We use machine learning to integrate diverse RNA sequence and structure features to model editing levels measured by deep sequencing. We confirm known features and identify new features important for RNA editing. Training and testing XGBoost algorithm within the same substrate yield models that explain 68 to 86 percent of substrate-specific variation in editing levels. However, the models do not generalize across substrates, suggesting complex and context-dependent regulation patterns. Our integrative approach can be applied to larger scale experiments towards deciphering the RNA editing code.


Subject(s)
Adenosine Deaminase/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Mutagenesis/genetics , RNA Editing/genetics , Regulatory Sequences, Nucleic Acid/genetics , Algorithms , Base Sequence , CRISPR-Associated Protein 9/metabolism , HEK293 Cells , Humans , Machine Learning , Models, Genetic , Mutation/genetics , Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , Substrate Specificity
8.
Nat Methods ; 17(7): 699-707, 2020 07.
Article in English | MEDLINE | ID: mdl-32616928

ABSTRACT

The discovery and design of biologically important RNA molecules is outpacing three-dimensional structural characterization. Here, we demonstrate that cryo-electron microscopy can routinely resolve maps of RNA-only systems and that these maps enable subnanometer-resolution coordinate estimation when complemented with multidimensional chemical mapping and Rosetta DRRAFTER computational modeling. This hybrid 'Ribosolve' pipeline detects and falsifies homologies and conformational rearrangements in 11 previously unknown 119- to 338-nucleotide protein-free RNA structures: full-length Tetrahymena ribozyme, hc16 ligase with and without substrate, full-length Vibrio cholerae and Fusobacterium nucleatum glycine riboswitch aptamers with and without glycine, Mycobacterium SAM-IV riboswitch with and without S-adenosylmethionine, and the computer-designed ATP-TTR-3 aptamer with and without AMP. Simulation benchmarks, blind challenges, compensatory mutagenesis, cross-RNA homologies and internal controls demonstrate that Ribosolve can accurately resolve the global architectures of RNA molecules but does not resolve atomic details. These tests offer guidelines for making inferences in future RNA structural studies with similarly accelerated throughput.


Subject(s)
Cryoelectron Microscopy/methods , RNA/chemistry , Computer Simulation , Models, Molecular , Nucleic Acid Conformation , RNA, Catalytic/chemistry , Riboswitch
9.
J Mol Biol ; 432(16): 4499-4522, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32512005

ABSTRACT

A hallmark of the initiation step of HIV-1 reverse transcription, in which viral RNA genome is converted into double-stranded DNA, is that it is slow and non-processive. Biochemical studies have identified specific sites along the viral RNA genomic template in which reverse transcriptase (RT) stalls. These stalling points, which occur after the addition of three and five template dNTPs, may serve as checkpoints to regulate the precise timing of HIV-1 reverse transcription following viral entry. Structural studies of reverse transcriptase initiation complexes (RTICs) have revealed unique conformations that may explain the slow rate of incorporation; however, questions remain about the temporal evolution of the complex and features that contribute to strong pausing during initiation. Here we present cryo-electron microscopy and single-molecule characterization of an RTIC after three rounds of dNTP incorporation (+3), the first major pausing point during reverse transcription initiation. Cryo-electron microscopy structures of a +3 extended RTIC reveal conformational heterogeneity within the RTIC core. Three distinct conformations were identified, two of which adopt unique, likely off-pathway, intermediates in the canonical polymerization cycle. Single-molecule Förster resonance energy transfer experiments confirm that the +3 RTIC is more structurally dynamic than earlier-stage RTICs. These alternative conformations were selectively disrupted through structure-guided point mutations to shift single-molecule Förster resonance energy transfer populations back toward the on-pathway conformation. Our results support the hypothesis that conformational heterogeneity within the HIV-1 RTIC during pausing serves as an additional means of regulating HIV-1 replication.


Subject(s)
DNA, Viral/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , Point Mutation , Cryoelectron Microscopy , DNA, Viral/metabolism , Fluorescence Resonance Energy Transfer , HIV Reverse Transcriptase/chemistry , HIV-1/metabolism , Models, Molecular , Molecular Conformation , Reverse Transcription , Single Molecule Imaging
10.
Nat Methods ; 17(7): 665-680, 2020 07.
Article in English | MEDLINE | ID: mdl-32483333

ABSTRACT

The Rosetta software for macromolecular modeling, docking and design is extensively used in laboratories worldwide. During two decades of development by a community of laboratories at more than 60 institutions, Rosetta has been continuously refactored and extended. Its advantages are its performance and interoperability between broad modeling capabilities. Here we review tools developed in the last 5 years, including over 80 methods. We discuss improvements to the score function, user interfaces and usability. Rosetta is available at http://www.rosettacommons.org.


Subject(s)
Macromolecular Substances/chemistry , Models, Molecular , Proteins/chemistry , Software , Molecular Docking Simulation , Peptidomimetics/chemistry , Protein Conformation
11.
Nat Commun ; 10(1): 5511, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796736

ABSTRACT

Specimens below 50 kDa have generally been considered too small to be analyzed by single-particle cryo-electron microscopy (cryo-EM). The high flexibility of pure RNAs makes it difficult to obtain high-resolution structures by cryo-EM. In bacteria, riboswitches regulate sulfur metabolism through binding to the S-adenosylmethionine (SAM) ligand and offer compelling targets for new antibiotics. SAM-I, SAM-I/IV, and SAM-IV are the three most commonly found SAM riboswitches, but the structure of SAM-IV is still unknown. Here, we report the structures of apo and SAM-bound SAM-IV riboswitches (119-nt, ~40 kDa) to 3.7 Å and 4.1 Å resolution, respectively, using cryo-EM. The structures illustrate homologies in the ligand-binding core but distinct peripheral tertiary contacts in SAM-IV compared to SAM-I and SAM-I/IV. Our results demonstrate the feasibility of resolving small RNAs with enough detail to enable detection of their ligand-binding pockets and suggest that cryo-EM could play a role in structure-assisted drug design for RNA.


Subject(s)
Cryoelectron Microscopy , Riboswitch , Ligands , Molecular Weight , Nucleic Acid Conformation , S-Adenosylmethionine/chemistry
12.
BMC Bioinformatics ; 20(1): 512, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31640563

ABSTRACT

BACKGROUND: The understanding of the importance of RNA has dramatically changed over recent years. As in the case of proteins, the function of an RNA molecule is encoded in its tertiary structure, which in turn is determined by the molecule's sequence. The prediction of tertiary structures of complex RNAs is still a challenging task. RESULTS: Using the observation that RNA sequences from the same RNA family fold into conserved structure, we test herein whether parallel modeling of RNA homologs can improve ab initio RNA structure prediction. EvoClustRNA is a multi-step modeling process, in which homologous sequences for the target sequence are selected using the Rfam database. Subsequently, independent folding simulations using Rosetta FARFAR and SimRNA are carried out. The model of the target sequence is selected based on the most common structural arrangement of the common helical fragments. As a test, on two blind RNA-Puzzles challenges, EvoClustRNA predictions ranked as the first of all submissions for the L-glutamine riboswitch and as the second for the ZMP riboswitch. Moreover, through a benchmark of known structures, we discovered several cases in which particular homologs were unusually amenable to structure recovery in folding simulations compared to the single original target sequence. CONCLUSION: This work, for the first time to our knowledge, demonstrates the importance of the selection of the target sequence from an alignment of an RNA family for the success of RNA 3D structure prediction. These observations prompt investigations into a new direction of research for checking 3D structure "foldability" or "predictability" of related RNA sequences to obtain accurate predictions. To support new research in this area, we provide all relevant scripts in a documented and ready-to-use form. By exploring new ideas and identifying limitations of the current RNA 3D structure prediction methods, this work is bringing us closer to the near-native computational RNA 3D models.


Subject(s)
Models, Molecular , RNA Folding , RNA/chemistry , Sequence Homology , Algorithms , Riboswitch , Software
13.
Nature ; 573(7774): 375-380, 2019 09.
Article in English | MEDLINE | ID: mdl-31485080

ABSTRACT

The molecular mechanisms of exon definition and back-splicing are fundamental unanswered questions in pre-mRNA splicing. Here we report cryo-electron microscopy structures of the yeast spliceosomal E complex assembled on introns, providing a view of the earliest event in the splicing cycle that commits pre-mRNAs to splicing. The E complex architecture suggests that the same spliceosome can assemble across an exon, and that it either remodels to span an intron for canonical linear splicing (typically on short exons) or catalyses back-splicing to generate circular RNA (on long exons). The model is supported by our experiments, which show that an E complex assembled on the middle exon of yeast EFM5 or HMRA1 can be chased into circular RNA when the exon is sufficiently long. This simple model unifies intron definition, exon definition, and back-splicing through the same spliceosome in all eukaryotes and should inspire experiments in many other systems to understand the mechanism and regulation of these processes.


Subject(s)
Exons , Introns , Models, Molecular , RNA Splicing , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cryoelectron Microscopy , Protein Structure, Quaternary , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Spliceosomes/metabolism , Spliceosomes/ultrastructure
14.
Mol Cell ; 74(5): 966-981.e18, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31078383

ABSTRACT

High-throughput methodologies have enabled routine generation of RNA target sets and sequence motifs for RNA-binding proteins (RBPs). Nevertheless, quantitative approaches are needed to capture the landscape of RNA-RBP interactions responsible for cellular regulation. We have used the RNA-MaP platform to directly measure equilibrium binding for thousands of designed RNAs and to construct a predictive model for RNA recognition by the human Pumilio proteins PUM1 and PUM2. Despite prior findings of linear sequence motifs, our measurements revealed widespread residue flipping and instances of positional coupling. Application of our thermodynamic model to published in vivo crosslinking data reveals quantitative agreement between predicted affinities and in vivo occupancies. Our analyses suggest a thermodynamically driven, continuous Pumilio-binding landscape that is negligibly affected by RNA structure or kinetic factors, such as displacement by ribosomes. This work provides a quantitative foundation for dissecting the cellular behavior of RBPs and cellular features that impact their occupancies.


Subject(s)
Nucleic Acid Conformation , RNA-Binding Proteins/genetics , Amino Acid Sequence/genetics , Humans , Kinetics , Protein Binding/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , Ribosomes/chemistry , Ribosomes/genetics
15.
Proc Natl Acad Sci U S A ; 116(17): 8336-8341, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30962376

ABSTRACT

Interactions between RNA and proteins are pervasive in biology, driving fundamental processes such as protein translation and participating in the regulation of gene expression. Modeling the energies of RNA-protein interactions is therefore critical for understanding and repurposing living systems but has been hindered by complexities unique to RNA-protein binding. Here, we bring together several advances to complete a calculation framework for RNA-protein binding affinities, including a unified free energy function for bound complexes, automated Rosetta modeling of mutations, and use of secondary structure-based energetic calculations to model unbound RNA states. The resulting Rosetta-Vienna RNP-ΔΔG method achieves root-mean-squared errors (RMSEs) of 1.3 kcal/mol on high-throughput MS2 coat protein-RNA measurements and 1.5 kcal/mol on an independent test set involving the signal recognition particle, human U1A, PUM1, and FOX-1. As a stringent test, the method achieves RMSE accuracy of 1.4 kcal/mol in blind predictions of hundreds of human PUM2-RNA relative binding affinities. Overall, these RMSE accuracies are significantly better than those attained by prior structure-based approaches applied to the same systems. Importantly, Rosetta-Vienna RNP-ΔΔG establishes a framework for further improvements in modeling RNA-protein binding that can be tested by prospective high-throughput measurements on new systems.


Subject(s)
Protein Binding , Protein Conformation , RNA-Binding Proteins , RNA , Binding Sites , Humans , Mutation , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Thermodynamics
16.
Structure ; 27(1): 140-151.e5, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30416038

ABSTRACT

RNA-protein complexes underlie numerous cellular processes including translation, splicing, and posttranscriptional regulation of gene expression. The structures of these complexes are crucial to their functions but often elude high-resolution structure determination. Computational methods are needed that can integrate low-resolution data for RNA-protein complexes while modeling de novo the large conformational changes of RNA components upon complex formation. To address this challenge, we describe RNP-denovo, a Rosetta method to simultaneously fold-and-dock RNA to a protein surface. On a benchmark set of diverse RNA-protein complexes not solvable with prior strategies, RNP-denovo consistently sampled native-like structures with better than nucleotide resolution. We revisited three past blind modeling challenges involving the spliceosome, telomerase, and a methyltransferase-ribosomal RNA complex in which previous methods gave poor results. When coupled with the same sparse FRET, crosslinking, and functional data used previously, RNP-denovo gave models with significantly improved accuracy. These results open a route to modeling global folds of RNA-protein complexes from low-resolution data.


Subject(s)
Molecular Docking Simulation/methods , Protein Domains , RNA/chemistry , Software , Binding Sites , Protein Binding , Protein Folding , Proteins/chemistry , Proteins/metabolism , RNA/metabolism , RNA Folding
17.
Nat Methods ; 15(11): 947-954, 2018 11.
Article in English | MEDLINE | ID: mdl-30377372

ABSTRACT

Increasingly, cryo-electron microscopy (cryo-EM) is used to determine the structures of RNA-protein assemblies, but nearly all maps determined with this method have biologically important regions where the local resolution does not permit RNA coordinate tracing. To address these omissions, we present de novo ribonucleoprotein modeling in real space through assembly of fragments together with experimental density in Rosetta (DRRAFTER). We show that DRRAFTER recovers near-native models for a diverse benchmark set of RNA-protein complexes including the spliceosome, mitochondrial ribosome, and CRISPR-Cas9-sgRNA complexes; rigorous blind tests include yeast U1 snRNP and spliceosomal P complex maps. Additionally, to aid in model interpretation, we present a method for reliable in situ estimation of DRRAFTER model accuracy. Finally, we apply DRRAFTER to recently determined maps of telomerase, the HIV-1 reverse transcriptase initiation complex, and the packaged MS2 genome, demonstrating the acceleration of accurate model building in challenging cases.


Subject(s)
Computational Biology/methods , Cryoelectron Microscopy/methods , Models, Molecular , RNA/ultrastructure , Ribonucleoproteins/ultrastructure , Software , Algorithms , Humans , Protein Conformation , RNA/metabolism , Ribonucleoproteins/metabolism
18.
Nature ; 557(7703): 118-122, 2018 05.
Article in English | MEDLINE | ID: mdl-29695867

ABSTRACT

Reverse transcription of the HIV-1 RNA genome into double-stranded DNA is a central step in viral infection 1 and a common target of antiretroviral drugs 2 . The reaction is catalysed by viral reverse transcriptase (RT)3,4 that is packaged in an infectious virion with two copies of viral genomic RNA 5 each bound to host lysine 3 transfer RNA (tRNALys3), which acts as a primer for initiation of reverse transcription6,7. Upon viral entry into cells, initiation is slow and non-processive compared to elongation8,9. Despite extensive efforts, the structural basis of RT function during initiation has remained a mystery. Here we use cryo-electron microscopy to determine a three-dimensional structure of an HIV-1 RT initiation complex. In our structure, RT is in an inactive polymerase conformation with open fingers and thumb and with the nucleic acid primer-template complex shifted away from the active site. The primer binding site (PBS) helix formed between tRNALys3 and HIV-1 RNA lies in the cleft of RT and is extended by additional pairing interactions. The 5' end of the tRNA refolds and stacks on the PBS to create a long helical structure, while the remaining viral RNA forms two helical stems positioned above the RT active site, with a linker that connects these helices to the RNase H region of the PBS. Our results illustrate how RNA structure in the initiation complex alters RT conformation to decrease activity, highlighting a potential target for drug action.


Subject(s)
Cryoelectron Microscopy , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/ultrastructure , HIV-1/enzymology , Base Sequence , Catalytic Domain , HIV Reverse Transcriptase/metabolism , Models, Molecular , Molecular Conformation , RNA, Transfer, Lys/chemistry , RNA, Transfer, Lys/metabolism , RNA, Transfer, Lys/ultrastructure , Reverse Transcription , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Ribonuclease H/ultrastructure
19.
J Chem Theory Comput ; 13(6): 3031-3048, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28430426

ABSTRACT

Over the past decade, the Rosetta biomolecular modeling suite has informed diverse biological questions and engineering challenges ranging from interpretation of low-resolution structural data to design of nanomaterials, protein therapeutics, and vaccines. Central to Rosetta's success is the energy function: a model parametrized from small-molecule and X-ray crystal structure data used to approximate the energy associated with each biomolecule conformation. This paper describes the mathematical models and physical concepts that underlie the latest Rosetta energy function, called the Rosetta Energy Function 2015 (REF15). Applying these concepts, we explain how to use Rosetta energies to identify and analyze the features of biomolecular models. Finally, we discuss the latest advances in the energy function that extend its capabilities from soluble proteins to also include membrane proteins, peptides containing noncanonical amino acids, small molecules, carbohydrates, nucleic acids, and other macromolecules.


Subject(s)
Macromolecular Substances/chemistry , Molecular Dynamics Simulation , HIV Protease/chemistry , HIV Protease/genetics , HIV Protease/metabolism , Macromolecular Substances/metabolism , Mutation , Protein Conformation , Static Electricity , Thermodynamics
20.
RNA ; 23(5): 655-672, 2017 05.
Article in English | MEDLINE | ID: mdl-28138060

ABSTRACT

RNA-Puzzles is a collective experiment in blind 3D RNA structure prediction. We report here a third round of RNA-Puzzles. Five puzzles, 4, 8, 12, 13, 14, all structures of riboswitch aptamers and puzzle 7, a ribozyme structure, are included in this round of the experiment. The riboswitch structures include biological binding sites for small molecules (S-adenosyl methionine, cyclic diadenosine monophosphate, 5-amino 4-imidazole carboxamide riboside 5'-triphosphate, glutamine) and proteins (YbxF), and one set describes large conformational changes between ligand-free and ligand-bound states. The Varkud satellite ribozyme is the most recently solved structure of a known large ribozyme. All puzzles have established biological functions and require structural understanding to appreciate their molecular mechanisms. Through the use of fast-track experimental data, including multidimensional chemical mapping, and accurate prediction of RNA secondary structure, a large portion of the contacts in 3D have been predicted correctly leading to similar topologies for the top ranking predictions. Template-based and homology-derived predictions could predict structures to particularly high accuracies. However, achieving biological insights from de novo prediction of RNA 3D structures still depends on the size and complexity of the RNA. Blind computational predictions of RNA structures already appear to provide useful structural information in many cases. Similar to the previous RNA-Puzzles Round II experiment, the prediction of non-Watson-Crick interactions and the observed high atomic clash scores reveal a notable need for an algorithm of improvement. All prediction models and assessment results are available at http://ahsoka.u-strasbg.fr/rnapuzzles/.


Subject(s)
RNA, Catalytic/chemistry , Riboswitch , Aminoimidazole Carboxamide/chemistry , Aminoimidazole Carboxamide/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Dinucleoside Phosphates/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Glutamine/chemistry , Glutamine/metabolism , Ligands , Models, Molecular , Nucleic Acid Conformation , RNA, Catalytic/metabolism , Ribonucleotides/chemistry , Ribonucleotides/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL