LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 4 of total 4

Search options

  1. Article ; Online: Inverse Mixed-Solvent Molecular Dynamics for Visualization of the Residue Interaction Profile of Molecular Probes

    Keisuke Yanagisawa / Ryunosuke Yoshino / Genki Kudo / Takatsugu Hirokawa

    International Journal of Molecular Sciences, Vol 23, Iss 4749, p

    2022  Volume 4749

    Abstract: To ensure efficiency in discovery and development, the application of computational technology is essential. Although virtual screening techniques are widely applied in the early stages of drug discovery research, the computational methods used in lead ... ...

    Abstract To ensure efficiency in discovery and development, the application of computational technology is essential. Although virtual screening techniques are widely applied in the early stages of drug discovery research, the computational methods used in lead optimization to improve activity and reduce the toxicity of compounds are still evolving. In this study, we propose a method to construct the residue interaction profile of the chemical structure used in the lead optimization by performing “inverse” mixed-solvent molecular dynamics (MSMD) simulation. Contrary to constructing a protein-based, atom interaction profile, we constructed a probe-based, protein residue interaction profile using MSMD trajectories. It provides us the profile of the preferred protein environments of probes without co-crystallized structures. We assessed the method using three probes: benzamidine, catechol, and benzene. As a result, the residue interaction profile of each probe obtained by MSMD was a reasonable physicochemical description of the general non-covalent interaction. Moreover, comparison with the X-ray structure containing each probe as a ligand shows that the map of the interaction profile matches the arrangement of amino acid residues in the X-ray structure.
    Keywords mixed-solvent molecular dynamics ; interaction pattern analysis ; residue interaction profile ; visualization ; Biology (General) ; QH301-705.5 ; Chemistry ; QD1-999
    Subject code 541
    Language English
    Publishing date 2022-04-01T00:00:00Z
    Publisher MDPI AG
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  2. Article ; Online: Solving Generalized Polyomino Puzzles Using the Ising Model

    Kazuki Takabatake / Keisuke Yanagisawa / Yutaka Akiyama

    Entropy, Vol 24, Iss 354, p

    2022  Volume 354

    Abstract: In the polyomino puzzle, the aim is to fill a finite space using several polyomino pieces with no overlaps or blanks. Because it is an NP-complete combinatorial optimization problem, various probabilistic and approximated approaches have been applied to ... ...

    Abstract In the polyomino puzzle, the aim is to fill a finite space using several polyomino pieces with no overlaps or blanks. Because it is an NP-complete combinatorial optimization problem, various probabilistic and approximated approaches have been applied to find solutions. Several previous studies embedded the polyomino puzzle in a QUBO problem, where the original objective function and constraints are transformed into the Hamiltonian function of the simulated Ising model. A solution to the puzzle is obtained by searching for a ground state of Hamiltonian by simulating the dynamics of the multiple-spin system. However, previous methods could solve only tiny polyomino puzzles considering a few combinations because their Hamiltonian designs were not efficient. We propose an improved Hamiltonian design that introduces new constraints and guiding terms to weakly encourage favorable spins and pairs in the early stages of computation. The proposed model solves the pentomino puzzle represented by approximately 2000 spins with >90% probability. Additionally, we extended the method to a generalized problem where each polyomino piece could be used zero or more times and solved it with approximately 100% probability. The proposed method also appeared to be effective for the 3D polycube puzzle, which is similar to applications in fragment-based drug discovery.
    Keywords Ising model ; polyomino puzzle ; Hopfield neural network ; combinatorial optimization ; Science ; Q ; Astrophysics ; QB460-466 ; Physics ; QC1-999
    Subject code 006
    Language English
    Publishing date 2022-02-01T00:00:00Z
    Publisher MDPI AG
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  3. Article ; Online: MEGADOCK-Web

    Takanori Hayashi / Yuri Matsuzaki / Keisuke Yanagisawa / Masahito Ohue / Yutaka Akiyama

    BMC Bioinformatics, Vol 19, Iss S4, Pp 61-

    an integrated database of high-throughput structure-based protein-protein interaction predictions

    2018  Volume 72

    Abstract: Abstract Background Protein-protein interactions (PPIs) play several roles in living cells, and computational PPI prediction is a major focus of many researchers. The three-dimensional (3D) structure and binding surface are important for the design of ... ...

    Abstract Abstract Background Protein-protein interactions (PPIs) play several roles in living cells, and computational PPI prediction is a major focus of many researchers. The three-dimensional (3D) structure and binding surface are important for the design of PPI inhibitors. Therefore, rigid body protein-protein docking calculations for two protein structures are expected to allow elucidation of PPIs different from known complexes in terms of 3D structures because known PPI information is not explicitly required. We have developed rapid PPI prediction software based on protein-protein docking, called MEGADOCK. In order to fully utilize the benefits of computational PPI predictions, it is necessary to construct a comprehensive database to gather prediction results and their predicted 3D complex structures and to make them easily accessible. Although several databases exist that provide predicted PPIs, the previous databases do not contain a sufficient number of entries for the purpose of discovering novel PPIs. Results In this study, we constructed an integrated database of MEGADOCK PPI predictions, named MEGADOCK-Web. MEGADOCK-Web provides more than 10 times the number of PPI predictions than previous databases and enables users to conduct PPI predictions that cannot be found in conventional PPI prediction databases. In MEGADOCK-Web, there are 7528 protein chains and 28,331,628 predicted PPIs from all possible combinations of those proteins. Each protein structure is annotated with PDB ID, chain ID, UniProt AC, related KEGG pathway IDs, and known PPI pairs. Additionally, MEGADOCK-Web provides four powerful functions: 1) searching precalculated PPI predictions, 2) providing annotations for each predicted protein pair with an experimentally known PPI, 3) visualizing candidates that may interact with the query protein on biochemical pathways, and 4) visualizing predicted complex structures through a 3D molecular viewer. Conclusion MEGADOCK-Web provides a huge amount of comprehensive PPI predictions based on docking ...
    Keywords Protein-protein interaction ; Protein-protein docking ; Predicted protein-protein interaction database ; MEGADOCK ; MEGADOCK-Web ; Computer applications to medicine. Medical informatics ; R858-859.7 ; Biology (General) ; QH301-705.5
    Subject code 612
    Language English
    Publishing date 2018-05-01T00:00:00Z
    Publisher BMC
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  4. Article ; Online: Computational prediction of plasma protein binding of cyclic peptides from small molecule experimental data using sparse modeling techniques

    Takashi Tajimi / Naoki Wakui / Keisuke Yanagisawa / Yasushi Yoshikawa / Masahito Ohue / Yutaka Akiyama

    BMC Bioinformatics, Vol 19, Iss S19, Pp 157-

    2018  Volume 170

    Abstract: Abstract Background Cyclic peptide-based drug discovery is attracting increasing interest owing to its potential to avoid target protein depletion. In drug discovery, it is important to maintain the biostability of a drug within the proper range. Plasma ... ...

    Abstract Abstract Background Cyclic peptide-based drug discovery is attracting increasing interest owing to its potential to avoid target protein depletion. In drug discovery, it is important to maintain the biostability of a drug within the proper range. Plasma protein binding (PPB) is the most important index of biostability, and developing a computational method to predict PPB of drug candidate compounds contributes to the acceleration of drug discovery research. PPB prediction of small molecule drug compounds using machine learning has been conducted thus far; however, no study has investigated cyclic peptides because experimental information of cyclic peptides is scarce. Results First, we adopted sparse modeling and small molecule information to construct a PPB prediction model for cyclic peptides. As cyclic peptide data are limited, applying multidimensional nonlinear models involves concerns regarding overfitting. However, models constructed by sparse modeling can avoid overfitting, offering high generalization performance and interpretability. More than 1000 PPB data of small molecules are available, and we used them to construct a prediction models with two enumeration methods: enumerating lasso solutions (ELS) and forward beam search (FBS). The accuracies of the prediction models constructed by ELS and FBS were equal to or better than those of conventional non-linear models (MAE = 0.167–0.174) on cross-validation of a small molecule compound dataset. Moreover, we showed that the prediction accuracies for cyclic peptides were close to those for small molecule compounds (MAE = 0.194–0.288). Such high accuracy could not be obtained by a simple method of learning from cyclic peptide data directly by lasso regression (MAE = 0.286–0.671) or ridge regression (MAE = 0.244–0.354). Conclusion In this study, we proposed a machine learning techniques that uses low-dimensional sparse modeling to predict the PPB value of cyclic peptides computationally. The low-dimensional sparse model not only exhibits excellent generalization performance but also improves interpretation of the prediction model. This can provide common an noteworthy knowledge for future cyclic peptide drug discovery studies.
    Keywords Sparse modeling ; Feature selection ; Cyclic peptide ; Biostability ; Plasma protein binding (PPB) ; Computer applications to medicine. Medical informatics ; R858-859.7 ; Biology (General) ; QH301-705.5
    Subject code 540
    Language English
    Publishing date 2018-12-01T00:00:00Z
    Publisher BMC
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

To top