scholarly journals Theoretical and computational modeling of naturally and artificially modified RNA nucleotides

2021 ◽  
Author(s):  
◽  
Travis Caleb Hurst

Ribonucleic acid (RNA) is a polymeric nucleic acid that is crucial for cellular function, regulating gene expression and encoding/decoding protein/DNA molecules. Recent discoveries of diverse functionality in non-coding RNAs have led to unprecedented demand for RNA 3D structure determination. With current technology, general, accurate prediction of 3D structures for large RNAs from the sequence remains computationally intractable. One of the principal challenges arises from the conformational flexibility of RNA, especially in loop/junction regions, which results in a rugged energy landscape. Several strategies exist to overcome this challenge, including incorporation of efficient experimental information and coarse-grained (CG) modeling to improve computational sampling of the structural ensemble. A second challenge is the inclusion of naturally modified derivatives of canonical RNA nucleotides in structure analysis. Most RNA prediction strategies rely upon the canonical nucleotides (adenine (A), uracil (U), guanine (G), and cytosine (C)), ignoring the effects of modified nucleotides on the structure and system dynamics. In general, RNA molecules contain rigid and flexible structural elements, which can be probed using efficient selective 2'-hydroxyl analyzed by primer extension (SHAPE) experiments. SHAPE experiments selectively modify flexible RNA nucleotides and can be processed to produce a characteristic reactivity profile for an RNA molecule that contains structural information. Incorporation of efficient experimental information, such as SHAPE, in predicting RNA 3D structure is highly desirable for overcoming the current knowledge gap between RNA sequence and 3D structure. In the first project, we introduce a physics-based model, the 3D structure-SHAPE relationship (3DSSR) model, to predict the SHAPE reactivity from the structure and show how this model may be used to sieve SHAPE-compatible structures from a pool of low-energy decoys and refine our predictions. In the second project, we compare 3DSSR performance to that of a convolutional neural network (CNN) trained on the SHAPE data and RNA structures, showing that 3DSSR outperforms the CNN given the limited data available. In the third project, we further improve the 3DSSR model, gaining deeper insights into the SHAPE reaction and biases. In the fourth project, we explore the theory underpinning the iterative simulated CG RNA folding model (IsRNA). In establishing the underlying mechanics driving the success of the model, we were able to clarify and improve the parameterization method while expanding the model interpretation, which should broaden application of the method to other biopolymers, such as protein. We found that the parameterization method follows statistical mechanics principles but also has a Bayesian interpretation. Further, we found that the parameterization process can benefit from application of the principle of maximum entropy, which improves simulation and parameterization efficiency. In the fifth project, we investigate the impact of nucleotide modification on the structure and configurational ensemble of RNA molecules using free energy calculations. By applying modifications to a common RNA hairpin, we estimate the impact on the stability of the structural ensemble, identifying specific interactions that drive changes to the potential of mean force (PMF) and showing the context and modification-dependence of the variable alterations to the structure stability.

2017 ◽  
Vol 33 (16) ◽  
pp. 2479-2486 ◽  
Author(s):  
Mélanie Boudard ◽  
Dominique Barth ◽  
Julie Bernauer ◽  
Alain Denise ◽  
Johanne Cohen

2019 ◽  
Vol 35 (21) ◽  
pp. 4459-4461 ◽  
Author(s):  
Sha Gong ◽  
Chengxin Zhang ◽  
Yang Zhang

Abstract Motivation Comparison of RNA 3D structures can be used to infer functional relationship of RNA molecules. Most of the current RNA structure alignment programs are built on size-dependent scales, which complicate the interpretation of structure and functional relations. Meanwhile, the low speed prevents the programs from being applied to large-scale RNA structural database search. Results We developed an open-source algorithm, RNA-align, for RNA 3D structure alignment which has the structure similarity scaled by a size-independent and statistically interpretable scoring metric. Large-scale benchmark tests show that RNA-align significantly outperforms other state-of-the-art programs in both alignment accuracy and running speed. The major advantage of RNA-align lies at the quick convergence of the heuristic alignment iterations and the coarse-grained secondary structure assignment, both of which are crucial to the speed and accuracy of RNA structure alignments. Availability and implementation https://zhanglab.ccmb.med.umich.edu/RNA-align/. Supplementary information Supplementary data are available at Bioinformatics online.


2019 ◽  
Vol 116 (42) ◽  
pp. 21022-21030 ◽  
Author(s):  
Hung T. Nguyen ◽  
Naoto Hori ◽  
D. Thirumalai

RNA molecules cannot fold in the absence of counterions. Experiments are typically performed in the presence of monovalent and divalent cations. How to treat the impact of a solution containing a mixture of both ion types on RNA folding has remained a challenging problem for decades. By exploiting the large concentration difference between divalent and monovalent ions used in experiments, we develop a theory based on the reference interaction site model (RISM), which allows us to treat divalent cations explicitly while keeping the implicit screening effect due to monovalent ions. Our theory captures both the inner shell and outer shell coordination of divalent cations to phosphate groups, which we demonstrate is crucial for an accurate calculation of RNA folding thermodynamics. The RISM theory for ion–phosphate interactions when combined with simulations based on a transferable coarse-grained model allows us to predict accurately the folding of several RNA molecules in a mixture containing monovalent and divalent ions. The calculated folding free energies and ion-preferential coefficients for RNA molecules (pseudoknots, a fragment of the rRNA, and the aptamer domain of the adenine riboswitch) are in excellent agreement with experiments over a wide range of monovalent and divalent ion concentrations. Because the theory is general, it can be readily used to investigate ion and sequence effects on DNA properties.


Life ◽  
2021 ◽  
Vol 11 (11) ◽  
pp. 1230
Author(s):  
Ching-Ming Lin ◽  
Jay-How Yang ◽  
Hwei-Jen Lee ◽  
Yu-Pang Lin ◽  
Li-Ping Tsai ◽  
...  

Background: Cockayne syndrome (CS) is a rare form of dwarfism that is characterized by progressive premature aging. CS is typically caused by mutations in the excision repair cross-complementing protein group 6 (ERCC6) gene that encodes the CS group B (CSB) protein. Using whole exome sequencing, we recently identified a novel homozygous missense mutation (Leu536Trp) in CSB in a Taiwanese boy with CS. Since the current database (Varsome) interprets this variant as likely pathogenic, we utilized a bioinformatic tool to investigate the impact of Leu536Trp as well as two other variants (Arg453Ter, Asp532Gly) in similar articles on the CSB protein structure stability. Methods: We used iterative threading assembly refinement (I-TASSER) to generate a predictive 3D structure of CSB. We calculated the change of mutation energy after residues substitution on the protein stability using I-TASSER as well as the artificial intelligence program Alphafold. Results: The Asp532Gly variant destabilized both modeled structures, while the Leu536Trp variant showed no effect on I-TASSER’s model but destabilized the Alphafold’s modeled structure. Conclusions: We propose here the first case of CS associated with a novel homozygous missense mutation (Leu536Trp) in CSB. Furthermore, we suggest that the Asp532Gly and Leu536Trp variants are both pathogenic after bioinformatic analysis of protein stability.


2019 ◽  
Author(s):  
Hung T. Nguyen ◽  
Naoto Hori ◽  
D. Thirumalai

RNA molecules cannot fold in the absence of counter ions. Experiments are typically performed in the presence of monovalent and divalent cations. How to treat the impact of a solution containing a mixture of both ion types on RNA folding has remained a challenging problem for decades. By exploiting the large concentration difference between divalent and monovalent ions used in experiments, we develop a theory based on the Reference Interaction Site Model (RISM), which allows us to treat divalent cations explicitly while keeping the implicit screening effect due to monovalent ions. Our theory captures both the inner shell and outer shell coordination of divalent cations to phosphate groups, which we demonstrate is crucial in an accurate calculation of RNA folding thermodynamics. The RISM theory for ion-phosphate interactions when combined with simulations based on a transferable coarse-grained model allows us to accurately predict the folding of several RNA molecules in a mixture containing monovalent and divalent ions. The calculated folding free energies and ion-preferential coefficients for RNA molecules (pseudoknots, a fragment of the ribosomal RNA, and the aptamer domain of the adenine riboswitch) are in excellent agreement with experiments over a wide range of monovalent and divalent ion concentrations. Because the theory is general, it can be readily used to investigate ion and sequence effects on DNA properties.Significance StatementRNA molecules require ions to fold. The problem of how ions of differing sizes and valences drive the folding of RNA molecules is unsolved. Here, we take a major step in its solution by creating a method, based on the theory of polyatomic liquids, to calculate the potential between divalent ions and the phosphate groups. The resulting model, accounting for inner and outer sphere coordination of Mg2+ and Ca2+ to phosphates, when used in coarse-grained molecular simulations predicts folding free energies for a number of RNA molecules in the presence of both divalent and monovalent ions that are in excellent agreement with experiments. The work sets the stage for probing sequence and ion effects on DNA and synthetic polyelectrolytes.


2019 ◽  
Vol 151 (16) ◽  
pp. 165101
Author(s):  
Ben-Gong Zhang ◽  
Hua-Hai Qiu ◽  
Jian Jiang ◽  
Jie Liu ◽  
Ya-Zhou Shi

2021 ◽  
Vol 8 ◽  
Author(s):  
Jun Li ◽  
Shi-Jie Chen

The three-dimensional (3D) structures of Ribonucleic acid (RNA) molecules are essential to understanding their various and important biological functions. However, experimental determination of the atomic structures is laborious and technically difficult. The large gap between the number of sequences and the experimentally determined structures enables the thriving development of computational approaches to modeling RNAs. However, computational methods based on all-atom simulations are intractable for large RNA systems, which demand long time simulations. Facing such a challenge, many coarse-grained (CG) models have been developed. Here, we provide a review of CG models for modeling RNA 3D structures, compare the performance of the different models, and offer insights into potential future developments.


2014 ◽  
Vol 141 (10) ◽  
pp. 105102 ◽  
Author(s):  
Ya-Zhou Shi ◽  
Feng-Hua Wang ◽  
Yuan-Yan Wu ◽  
Zhi-Jie Tan

2018 ◽  
Author(s):  
L. Jin ◽  
Y.Z. Shi ◽  
C.J. Feng ◽  
Y.L. Tan ◽  
Z.J. Tan

AbstractDouble-stranded (ds) RNAs play essential roles in many processes of cell metabolism. The knowledge of three-dimensional (3D) structure, stability and flexibility of dsRNAs in salt solutions is important for understanding their biological functions. In this work, we further developed our previously proposed coarse-grained model to predict 3D structure, stability and flexibility for dsRNAs in monovalent and divalent ion solutions through involving an implicit structure-based electrostatic potential. The model can make reliable predictions for 3D structures of extensive dsRNAs with/without bulge/internal loops from their sequences, and the involvement of the structure-based electrostatic potential and corresponding ion condition can improve the predictions on 3D structures of dsRNAs in ion solutions. Furthermore, the model can make good predictions on thermal stability for extensive dsRNAs over the wide range of monovalent/divalent ion concentrations, and our analyses show that thermally unfolding pathway of a dsRNA is generally dependent on its length as well as its sequence. In addition, the model was employed to examine the salt-dependent flexibility of a dsRNA helix and the calculated salt-dependent persistence lengths are in good accordance with experiments.


2019 ◽  
Author(s):  
Robin A. Corey ◽  
Owen N. Vickery ◽  
Mark S. P. Sansom ◽  
Phillip J. Stansfeld

AbstractIntegral membrane proteins are regulated by specific interactions with lipids from the surrounding bilayer. The structures of protein-lipid complexes can be determined through a combination of experimental and computational approaches, but the energetic basis of these interactions is difficult to resolve. Molecular dynamic simulations provide the primary computational technique to estimate the free energies of these interactions. We demonstrate that the energetics of protein-lipid interactions may be reliably and reproducibly calculated using three simulation-based approaches: potential of mean force calculations. alchemical free energy perturbation, and well-tempered metadynamics. We employ these techniques within the framework of a coarse-grained force field, and apply them to both bacterial and mammalian membrane protein-lipid systems. We demonstrate good agreement between the different techniques, providing a robust framework for their automated implementation within a pipeline for annotation of newly determined membrane protein structures.


Sign in / Sign up

Export Citation Format

Share Document