Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hisanori Kiryu is active.

Publication


Featured researches published by Hisanori Kiryu.


Bioinformatics | 2009

Prediction of RNA secondary structure using generalized centroid estimators

Michiaki Hamada; Hisanori Kiryu; Kengo Sato; Toutai Mituyama; Kiyoshi Asai

MOTIVATION Recent studies have shown that the methods for predicting secondary structures of RNAs on the basis of posterior decoding of the base-pairing probabilities has an advantage with respect to prediction accuracy over the conventionally utilized minimum free energy methods. However, there is room for improvement in the objective functions presented in previous studies, which are maximized in the posterior decoding with respect to the accuracy measures for secondary structures. RESULTS We propose novel estimators which improve the accuracy of secondary structure prediction of RNAs. The proposed estimators maximize an objective function which is the weighted sum of the expected number of the true positives and that of the true negatives of the base pairs. The proposed estimators are also improved versions of the ones used in previous works, namely CONTRAfold for secondary structure prediction from a single RNA sequence and McCaskill-MEA for common secondary structure prediction from multiple alignments of RNA sequences. We clarify the relations between the proposed estimators and the estimators presented in previous works, and theoretically show that the previous estimators include additional unnecessary terms in the evaluation measures with respect to the accuracy. Furthermore, computational experiments confirm the theoretical analysis by indicating improvement in the empirical accuracy. The proposed estimators represent extensions of the centroid estimators proposed in Ding et al. and Carvalho and Lawrence, and are applicable to a wide variety of problems in bioinformatics. AVAILABILITY Supporting information and the CentroidFold software are available online at: http://www.ncrna.org/software/centroidfold/.


BMC Bioinformatics | 2008

A fast structural multiple alignment method for long RNA sequences

Yasuo Tabei; Hisanori Kiryu; Taishin Kin; Kiyoshi Asai

BackgroundAligning multiple RNA sequences is essential for analyzing non-coding RNAs. Although many alignment methods for non-coding RNAs, including Sankoffs algorithm for strict structural alignments, have been proposed, they are either inaccurate or computationally too expensive. Faster methods with reasonable accuracies are required for genome-scale analyses.ResultsWe propose a fast algorithm for multiple structural alignments of RNA sequences that is an extension of our pairwise structural alignment method (implemented in SCARNA). The accuracies of the implemented software, MXSCARNA, are at least as favorable as those of state-of-art algorithms that are computationally much more expensive in time and memory.ConclusionThe proposed method for structural alignment of multiple RNA sequences is fast enough for large-scale analyses with accuracies at least comparable to those of existing algorithms. The source code of MXSCARNA and its web server are available at http://mxscarna.ncrna.org.


Bioinformatics | 2007

Robust prediction of consensus secondary structures using averaged base pairing probability matrices

Hisanori Kiryu; Taishin Kin; Kiyoshi Asai

MOTIVATION Recent transcriptomic studies have revealed the existence of a considerable number of non-protein-coding RNA transcripts in higher eukaryotic cells. To investigate the functional roles of these transcripts, it is of great interest to find conserved secondary structures from multiple alignments on a genomic scale. Since multiple alignments are often created using alignment programs that neglect the special conservation patterns of RNA secondary structures for computational efficiency, alignment failures can cause potential risks of overlooking conserved stem structures. RESULTS We investigated the dependence of the accuracy of secondary structure prediction on the quality of alignments. We compared three algorithms that maximize the expected accuracy of secondary structures as well as other frequently used algorithms. We found that one of our algorithms, called McCaskill-MEA, was more robust against alignment failures than others. The McCaskill-MEA method first computes the base pairing probability matrices for all the sequences in the alignment and then obtains the base pairing probability matrix of the alignment by averaging over these matrices. The consensus secondary structure is predicted from this matrix such that the expected accuracy of the prediction is maximized. We show that the McCaskill-MEA method performs better than other methods, particularly when the alignment quality is low and when the alignment consists of many sequences. Our model has a parameter that controls the sensitivity and specificity of predictions. We discussed the uses of that parameter for multi-step screening procedures to search for conserved secondary structures and for assigning confidence values to the predicted base pairs. AVAILABILITY The C++ source code that implements the McCaskill-MEA algorithm and the test dataset used in this paper are available at http://www.ncrna.org/papers/McCaskillMEA/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Bioinformatics | 2009

CentroidAlign: fast and accurate aligner for structured RNAs by maximizing expected sum-of-pairs score.

Michiaki Hamada; Kengo Sato; Hisanori Kiryu; Toutai Mituyama; Kiyoshi Asai

MOTIVATION The importance of accurate and fast predictions of multiple alignments for RNA sequences has increased due to recent findings about functional non-coding RNAs. Recent studies suggest that maximizing the expected accuracy of predictions will be useful for many problems in bioinformatics. RESULTS We designed a novel estimator for multiple alignments of structured RNAs, based on maximizing the expected accuracy of predictions. First, we define the maximum expected accuracy (MEA) estimator for pairwise alignment of RNA sequences. This maximizes the expected sum-of-pairs score (SPS) of a predicted alignment under a probability distribution of alignments given by marginalizing the Sankoff model. Then, by approximating the MEA estimator, we obtain an estimator whose time complexity is O(L(3)+c(2)dL(2)) where L is the length of input sequences and both c and d are constants independent of L. The proposed estimator can handle uncertainty of secondary structures and alignments that are obstacles in Bioinformatics because it considers all the secondary structures and all the pairwise alignments as input sequences. Moreover, we integrate the probabilistic consistency transformation (PCT) on alignments into the proposed estimator. Computational experiments using six benchmark datasets indicate that the proposed method achieved a favorable SPS and was the fastest of many state-of-the-art tools for multiple alignments of structured RNAs. AVAILABILITY The software called CentroidAlign, which is an implementation of the algorithm in this article, is freely available on our website: http://www.ncrna.org/software/centroidalign/. CONTACT [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Bioinformatics | 2009

Predictions of RNA secondary structure by combining homologous sequence information

Michiaki Hamada; Kengo Sato; Hisanori Kiryu; Toutai Mituyama; Kiyoshi Asai

Motivation: Secondary structure prediction of RNA sequences is an important problem. There have been progresses in this area, but the accuracy of prediction from an RNA sequence is still limited. In many cases, however, homologous RNA sequences are available with the target RNA sequence whose secondary structure is to be predicted. Results: In this article, we propose a new method for secondary structure predictions of individual RNA sequences by taking the information of their homologous sequences into account without assuming the common secondary structure of the entire sequences. The proposed method is based on posterior decoding techniques, which consider all the suboptimal secondary structures of the target and homologous sequences and all the suboptimal alignments between the target sequence and each of the homologous sequences. In our computational experiments, the proposed method provides better predictions than those performed only on the basis of the formation of individual RNA sequences and those performed by using methods for predicting the common secondary structure of the homologous sequences. Remarkably, we found that the common secondary predictions sometimes give worse predictions for the secondary structure of a target sequence than the predictions from the individual target sequence, while the proposed method always gives good predictions for the secondary structure of target sequences in all tested cases. Availability: Supporting information and software are available online at: http://www.ncrna.org/software/centroidfold/ismb2009/. Contact: [email protected] Supplementary information:Supplementary data are available at Bioinformatics online.


Genome Biology | 2014

CapR: revealing structural specificities of RNA-binding protein target recognition using CLIP-seq data

Tsukasa Fukunaga; Haruka Ozaki; Goro Terai; Kiyoshi Asai; Wataru Iwasaki; Hisanori Kiryu

RNA-binding proteins (RBPs) bind to their target RNA molecules by recognizing specific RNA sequences and structural contexts. The development of CLIP-seq and related protocols has made it possible to exhaustively identify RNA fragments that bind to RBPs. However, no efficient bioinformatics method exists to reveal the structural specificities of RBP–RNA interactions using these data. We present CapR, an efficient algorithm that calculates the probability that each RNA base position is located within each secondary structural context. Using CapR, we demonstrate that several RBPs bind to their target RNA molecules under specific structural contexts. CapR is available at https://sites.google.com/site/fukunagatsu/software/capr.


PLOS ONE | 2011

Generalized Centroid Estimators in Bioinformatics

Michiaki Hamada; Hisanori Kiryu; Wataru Iwasaki; Kiyoshi Asai

In a number of estimation problems in bioinformatics, accuracy measures of the target problem are usually given, and it is important to design estimators that are suitable to those accuracy measures. However, there is often a discrepancy between an employed estimator and a given accuracy measure of the problem. In this study, we introduce a general class of efficient estimators for estimation problems on high-dimensional binary spaces, which represent many fundamental problems in bioinformatics. Theoretical analysis reveals that the proposed estimators generally fit with commonly-used accuracy measures (e.g. sensitivity, PPV, MCC and F-score) as well as it can be computed efficiently in many cases, and cover a wide range of problems in bioinformatics from the viewpoint of the principle of maximum expected accuracy (MEA). It is also shown that some important algorithms in bioinformatics can be interpreted in a unified manner. Not only the concept presented in this paper gives a useful framework to design MEA-based estimators but also it is highly extendable and sheds new light on many problems in bioinformatics.


Bioinformatics | 2017

SCODE: An efficient regulatory network inference algorithm from single-cell RNA-Seq during differentiation

Hirotaka Matsumoto; Hisanori Kiryu; Chikara Furusawa; Minoru S.H. Ko; Shigeru B.H. Ko; Norio Gouda; Tetsutaro Hayashi; Itoshi Nikaido

Motivation: The analysis of RNA‐Seq data from individual differentiating cells enables us to reconstruct the differentiation process and the degree of differentiation (in pseudo‐time) of each cell. Such analyses can reveal detailed expression dynamics and functional relationships for differentiation. To further elucidate differentiation processes, more insight into gene regulatory networks is required. The pseudo‐time can be regarded as time information and, therefore, single‐cell RNA‐Seq data are time‐course data with high time resolution. Although time‐course data are useful for inferring networks, conventional inference algorithms for such data suffer from high time complexity when the number of samples and genes is large. Therefore, a novel algorithm is necessary to infer networks from single‐cell RNA‐Seq during differentiation. Results: In this study, we developed the novel and efficient algorithm SCODE to infer regulatory networks, based on ordinary differential equations. We applied SCODE to three single‐cell RNA‐Seq datasets and confirmed that SCODE can reconstruct observed expression dynamics. We evaluated SCODE by comparing its inferred networks with use of a DNaseI‐footprint based network. The performance of SCODE was best for two of the datasets and nearly best for the remaining dataset. We also compared the runtimes and showed that the runtimes for SCODE are significantly shorter than for alternatives. Thus, our algorithm provides a promising approach for further single‐cell differentiation analyses. Availability and Implementation: The R source code of SCODE is available at https://github.com/hmatsu1226/SCODE Contact: [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


BMC Bioinformatics | 2016

SCOUP: a probabilistic model based on the Ornstein–Uhlenbeck process to analyze single-cell expression data during differentiation

Hirotaka Matsumoto; Hisanori Kiryu

BackgroundSingle-cell technologies make it possible to quantify the comprehensive states of individual cells, and have the power to shed light on cellular differentiation in particular. Although several methods have been developed to fully analyze the single-cell expression data, there is still room for improvement in the analysis of differentiation.ResultsIn this paper, we propose a novel method SCOUP to elucidate differentiation process. Unlike previous dimension reduction-based approaches, SCOUP describes the dynamics of gene expression throughout differentiation directly, including the degree of differentiation of a cell (in pseudo-time) and cell fate. SCOUP is superior to previous methods with respect to pseudo-time estimation, especially for single-cell RNA-seq. SCOUP also successfully estimates cell lineage more accurately than previous method, especially for cells at an early stage of bifurcation. In addition, SCOUP can be applied to various downstream analyses. As an example, we propose a novel correlation calculation method for elucidating regulatory relationships among genes. We apply this method to a single-cell RNA-seq data and detect a candidate of key regulator for differentiation and clusters in a correlation network which are not detected with conventional correlation analysis.ConclusionsWe develop a stochastic process-based method SCOUP to analyze single-cell expression data throughout differentiation. SCOUP can estimate pseudo-time and cell lineage more accurately than previous methods. We also propose a novel correlation calculation method based on SCOUP. SCOUP is a promising approach for further single-cell analysis and available at https://github.com/hmatsu1226/SCOUP.


BMC Bioinformatics | 2016

Parallel computation of genome-scale RNA secondary structure to detect structural constraints on human genome

Risa Kawaguchi; Hisanori Kiryu

BackgroundRNA secondary structure around splice sites is known to assist normal splicing by promoting spliceosome recognition. However, analyzing the structural properties of entire intronic regions or pre-mRNA sequences has been difficult hitherto, owing to serious experimental and computational limitations, such as low read coverage and numerical problems.ResultsOur novel software, “ParasoR”, is designed to run on a computer cluster and enables the exact computation of various structural features of long RNA sequences under the constraint of maximal base-pairing distance. ParasoR divides dynamic programming (DP) matrices into smaller pieces, such that each piece can be computed by a separate computer node without losing the connectivity information between the pieces. ParasoR directly computes the ratios of DP variables to avoid the reduction of numerical precision caused by the cancellation of a large number of Boltzmann factors. The structural preferences of mRNAs computed by ParasoR shows a high concordance with those determined by high-throughput sequencing analyses.Using ParasoR, we investigated the global structural preferences of transcribed regions in the human genome. A genome-wide folding simulation indicated that transcribed regions are significantly more structural than intergenic regions after removing repeat sequences and k-mer frequency bias. In particular, we observed a highly significant preference for base pairing over entire intronic regions as compared to their antisense sequences, as well as to intergenic regions. A comparison between pre-mRNAs and mRNAs showed that coding regions become more accessible after splicing, indicating constraints for translational efficiency. Such changes are correlated with gene expression levels, as well as GC content, and are enriched among genes associated with cytoskeleton and kinase functions.ConclusionsWe have shown that ParasoR is very useful for analyzing the structural properties of long RNA sequences such as mRNAs, pre-mRNAs, and long non-coding RNAs whose lengths can be more than a million bases in the human genome. In our analyses, transcribed regions including introns are indicated to be subject to various types of structural constraints that cannot be explained from simple sequence composition biases. ParasoR is freely available at https://github.com/carushi/ParasoR.

Collaboration


Dive into the Hisanori Kiryu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Taishin Kin

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Toutai Mituyama

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Goro Terai

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yasuo Tabei

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Itoshi Nikaido

Yokohama City University

View shared research outputs
Researchain Logo
Decentralizing Knowledge