Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioinformatics ; 40(1)2024 01 02.
Article in English | MEDLINE | ID: mdl-38147362

ABSTRACT

MOTIVATION: Up-to-date pathway knowledge is usually presented in scientific publications for human reading, making it difficult to utilize these resources for semantic integration and computational analysis of biological pathways. We here present an approach to mining knowledge graphs by combining manual curation with automated named entity recognition and automated relation extraction. This approach allows us to study pathway-related questions in detail, which we here show using the ketamine pathway, aiming to help improve understanding of the role of gut microbiota in the antidepressant effects of ketamine. RESULTS: The thus devised ketamine pathway 'KetPath' knowledge graph comprises five parts: (i) manually curated pathway facts from images; (ii) recognized named entities in biomedical texts; (iii) identified relations between named entities; (iv) our previously constructed microbiota and pre-/probiotics knowledge bases; and (v) multiple community-accepted public databases. We first assessed the performance of automated extraction of relations between named entities using the specially designed state-of-the-art tool BioKetBERT. The query results show that we can retrieve drug actions, pathway relations, co-occurring entities, and their relations. These results uncover several biological findings, such as various gut microbes leading to increased expression of BDNF, which may contribute to the sustained antidepressant effects of ketamine. We envision that the methods and findings from this research will aid researchers who wish to integrate and query data and knowledge from multiple biomedical databases and literature simultaneously. AVAILABILITY AND IMPLEMENTATION: Data and query protocols are available in the KetPath repository at https://dx.doi.org/10.5281/zenodo.8398941 and https://github.com/tingcosmos/KetPath.


Subject(s)
Gastrointestinal Microbiome , Ketamine , Humans , Ketamine/pharmacology , Databases, Factual , Antidepressive Agents/pharmacology , Neurotransmitter Agents , Data Mining/methods
2.
Nat Rev Genet ; 20(11): 693-701, 2019 11.
Article in English | MEDLINE | ID: mdl-31455890

ABSTRACT

Human genomics is undergoing a step change from being a predominantly research-driven activity to one driven through health care as many countries in Europe now have nascent precision medicine programmes. To maximize the value of the genomic data generated, these data will need to be shared between institutions and across countries. In recognition of this challenge, 21 European countries recently signed a declaration to transnationally share data on at least 1 million human genomes by 2022. In this Roadmap, we identify the challenges of data sharing across borders and demonstrate that European research infrastructures are well-positioned to support the rapid implementation of widespread genomic data access.


Subject(s)
Biomedical Research , Genome, Human , Human Genome Project , Europe , Humans
4.
Bioinformatics ; 38(8): 2111-2118, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35150231

ABSTRACT

MOTIVATION: The interactions between proteins and other molecules are essential to many biological and cellular processes. Experimental identification of interface residues is a time-consuming, costly and challenging task, while protein sequence data are ubiquitous. Consequently, many computational and machine learning approaches have been developed over the years to predict such interface residues from sequence. However, the effectiveness of different Deep Learning (DL) architectures and learning strategies for protein-protein, protein-nucleotide and protein-small molecule interface prediction has not yet been investigated in great detail. Therefore, we here explore the prediction of protein interface residues using six DL architectures and various learning strategies with sequence-derived input features. RESULTS: We constructed a large dataset dubbed BioDL, comprising protein-protein interactions from the PDB, and DNA/RNA and small molecule interactions from the BioLip database. We also constructed six DL architectures, and evaluated them on the BioDL benchmarks. This shows that no single architecture performs best on all instances. An ensemble architecture, which combines all six architectures, does consistently achieve peak prediction accuracy. We confirmed these results on the published benchmark set by Zhang and Kurgan (ZK448), and on our own existing curated homo- and heteromeric protein interaction dataset. Our PIPENN sequence-based ensemble predictor outperforms current state-of-the-art sequence-based protein interface predictors on ZK448 on all interaction types, achieving an AUC-ROC of 0.718 for protein-protein, 0.823 for protein-nucleotide and 0.842 for protein-small molecule. AVAILABILITY AND IMPLEMENTATION: Source code and datasets are available at https://github.com/ibivu/pipenn/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Machine Learning , Proteins , Proteins/chemistry , Software , Amino Acid Sequence , Nucleotides , Computational Biology/methods
5.
Bioinformatics ; 37(20): 3421-3427, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-33974039

ABSTRACT

MOTIVATION: Antibodies play an important role in clinical research and biotechnology, with their specificity determined by the interaction with the antigen's epitope region, as a special type of protein-protein interaction (PPI) interface. The ubiquitous availability of sequence data, allows us to predict epitopes from sequence in order to focus time-consuming wet-lab experiments toward the most promising epitope regions. Here, we extend our previously developed sequence-based predictors for homodimer and heterodimer PPI interfaces to predict epitope residues that have the potential to bind an antibody. RESULTS: We collected and curated a high quality epitope dataset from the SAbDab database. Our generic PPI heterodimer predictor obtained an AUC-ROC of 0.666 when evaluated on the epitope test set. We then trained a random forest model specifically on the epitope dataset, reaching AUC 0.694. Further training on the combined heterodimer and epitope datasets, improves our final predictor to AUC 0.703 on the epitope test set. This is better than the best state-of-the-art sequence-based epitope predictor BepiPred-2.0. On one solved antibody-antigen structure of the COVID19 virus spike receptor binding domain, our predictor reaches AUC 0.778. We added the SeRenDIP-CE Conformational Epitope predictors to our webserver, which is simple to use and only requires a single antigen sequence as input, which will help make the method immediately applicable in a wide range of biomedical and biomolecular research. AVAILABILITY AND IMPLEMENTATION: Webserver, source code and datasets at www.ibi.vu.nl/programs/serendipwww/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

6.
Brief Bioinform ; 20(2): 540-550, 2019 03 22.
Article in English | MEDLINE | ID: mdl-28968694

ABSTRACT

This review provides a historical overview of the inception and development of bioinformatics research in the Netherlands. Rooted in theoretical biology by foundational figures such as Paulien Hogeweg (at Utrecht University since the 1970s), the developments leading to organizational structures supporting a relatively large Dutch bioinformatics community will be reviewed. We will show that the most valuable resource that we have built over these years is the close-knit national expert community that is well engaged in basic and translational life science research programmes. The Dutch bioinformatics community is accustomed to facing the ever-changing landscape of data challenges and working towards solutions together. In addition, this community is the stable factor on the road towards sustainability, especially in times where existing funding models are challenged and change rapidly.


Subject(s)
Community Networks , Computational Biology/methods , Computational Biology/organization & administration , Sequence Analysis, DNA/standards , Translational Research, Biomedical , Humans , Netherlands
7.
Bioinformatics ; 36(7): 2142-2149, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31845959

ABSTRACT

MOTIVATION: Genetic interaction (GI) patterns are characterized by the phenotypes of interacting single and double mutated gene pairs. Uncovering the regulatory mechanisms of GIs would provide a better understanding of their role in biological processes, diseases and drug response. Computational analyses can provide insights into the underpinning mechanisms of GIs. RESULTS: In this study, we present a framework for exhaustive modelling of GI patterns using Petri nets (PN). Four-node models were defined and generated on three levels with restrictions, to enable an exhaustive approach. Simulations suggest ∼5 million models of GIs. Generalizing these we propose putative mechanisms for the GI patterns, inversion and suppression. We demonstrate that exhaustive PN modelling enables reasoning about mechanisms of GIs when only the phenotypes of gene pairs are known. The framework can be applied to other GI or genetic regulatory datasets. AVAILABILITY AND IMPLEMENTATION: The framework is available at http://www.ibi.vu.nl/programs/ExhMod. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

8.
Bioinformatics ; 35(24): 5315-5317, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31368486

ABSTRACT

SUMMARY: PRALINE 2 is a toolkit for custom multiple sequence alignment workflows. It can be used to incorporate sequence annotations, such as secondary structure or (DNA) motifs, into the alignment scoring, as well as to customize many other aspects of a progressive multiple alignment workflow. AVAILABILITY AND IMPLEMENTATION: PRALINE 2 is implemented in Python and available as open source software on GitHub: https://github.com/ibivu/PRALINE/.


Subject(s)
Software , DNA , Protein Structure, Secondary , Sequence Alignment
9.
Bioinformatics ; 35(22): 4794-4796, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31116381

ABSTRACT

MOTIVATION: Interpretation of ubiquitous protein sequence data has become a bottleneck in biomolecular research, due to a lack of structural and other experimental annotation data for these proteins. Prediction of protein interaction sites from sequence may be a viable substitute. We therefore recently developed a sequence-based random forest method for protein-protein interface prediction, which yielded a significantly increased performance than other methods on both homomeric and heteromeric protein-protein interactions. Here, we present a webserver that implements this method efficiently. RESULTS: With the aim of accelerating our previous approach, we obtained sequence conservation profiles by re-mastering the alignment of homologous sequences found by PSI-BLAST. This yielded a more than 10-fold speedup and at least the same accuracy, as reported previously for our method; these results allowed us to offer the method as a webserver. The web-server interface is targeted to the non-expert user. The input is simply a sequence of the protein of interest, and the output a table with scores indicating the likelihood of having an interaction interface at a certain position. As the method is sequence-based and not sensitive to the type of protein interaction, we expect this webserver to be of interest to many biological researchers in academia and in industry. AVAILABILITY AND IMPLEMENTATION: Webserver, source code and datasets are available at www.ibi.vu.nl/programs/serendipwww/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Algorithms , Amino Acid Sequence , Proteins , Sequence Analysis, Protein
10.
PLoS Comput Biol ; 15(5): e1007061, 2019 05.
Article in English | MEDLINE | ID: mdl-31083661

ABSTRACT

Genetic interactions, a phenomenon whereby combinations of mutations lead to unexpected effects, reflect how cellular processes are wired and play an important role in complex genetic diseases. Understanding the molecular basis of genetic interactions is crucial for deciphering pathway organization as well as understanding the relationship between genetic variation and disease. Several hypothetical molecular mechanisms have been linked to different genetic interaction types. However, differences in genetic interaction patterns and their underlying mechanisms have not yet been compared systematically between different functional gene classes. Here, differences in the occurrence and types of genetic interactions are compared for two classes, gene-specific transcription factors (GSTFs) and signaling genes (kinases and phosphatases). Genome-wide gene expression data for 63 single and double deletion mutants in baker's yeast reveals that the two most common genetic interaction patterns are buffering and inversion. Buffering is typically associated with redundancy and is well understood. In inversion, genes show opposite behavior in the double mutant compared to the corresponding single mutants. The underlying mechanism is poorly understood. Although both classes show buffering and inversion patterns, the prevalence of inversion is much stronger in GSTFs. To decipher potential mechanisms, a Petri Net modeling approach was employed, where genes are represented as nodes and relationships between genes as edges. This allowed over 9 million possible three and four node models to be exhaustively enumerated. The models show that a quantitative difference in interaction strength is a strict requirement for obtaining inversion. In addition, this difference is frequently accompanied with a second gene that shows buffering. Taken together, these results provide a mechanistic explanation for inversion. Furthermore, the ability of transcription factors to differentially regulate expression of their targets provides a likely explanation why inversion is more prevalent for GSTFs compared to kinases and phosphatases.


Subject(s)
Gene Expression Regulation , Models, Genetic , Transcription Factors/metabolism , Chromosome Inversion , Computational Biology , Computer Simulation , Databases, Genetic , Epistasis, Genetic , Genes, Fungal , Genetic Association Studies , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Signal Transduction/genetics
11.
Bioinformatics ; 34(13): i4-i12, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29950011

ABSTRACT

Motivation: Our society has become data-rich to the extent that research in many areas has become impossible without computational approaches. Educational programmes seem to be lagging behind this development. At the same time, there is a growing need not only for strong data science skills, but foremost for the ability to both translate between tools and methods on the one hand, and application and problems on the other. Results: Here we present our experiences with shaping and running a masters' programme in bioinformatics and systems biology in Amsterdam. From this, we have developed a comprehensive philosophy on how translation in training may be achieved in a dynamic and multidisciplinary research area, which is described here. We furthermore describe two requirements that enable translation, which we have found to be crucial: sufficient depth and focus on multidisciplinary topic areas, coupled with a balanced breadth from adjacent disciplines. Finally, we present concrete suggestions on how this may be implemented in practice, which may be relevant for the effectiveness of life science and data science curricula in general, and of particular interest to those who are in the process of setting up such curricula. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Computational Biology/education , Curriculum , Data Science/education , Humans
12.
PLoS Comput Biol ; 14(11): e1006547, 2018 11.
Article in English | MEDLINE | ID: mdl-30383764

ABSTRACT

Protein or DNA motifs are sequence regions which possess biological importance. These regions are often highly conserved among homologous sequences. The generation of multiple sequence alignments (MSAs) with a correct alignment of the conserved sequence motifs is still difficult to achieve, due to the fact that the contribution of these typically short fragments is overshadowed by the rest of the sequence. Here we extended the PRALINE multiple sequence alignment program with a novel motif-aware MSA algorithm in order to address this shortcoming. This method can incorporate explicit information about the presence of externally provided sequence motifs, which is then used in the dynamic programming step by boosting the amino acid substitution matrix towards the motif. The strength of the boost is controlled by a parameter, α. Using a benchmark set of alignments we confirm that a good compromise can be found that improves the matching of motif regions while not significantly reducing the overall alignment quality. By estimating α on an unrelated set of reference alignments we find there is indeed a strong conservation signal for motifs. A number of typical but difficult MSA use cases are explored to exemplify the problems in correctly aligning functional sequence motifs and how the motif-aware alignment method can be employed to alleviate these problems.


Subject(s)
Amino Acid Motifs , DNA/chemistry , Proteins/chemistry , Sequence Alignment/standards , Algorithms , Amino Acid Sequence , Conserved Sequence , HIV-1/chemistry , Sequence Homology, Amino Acid , env Gene Products, Human Immunodeficiency Virus/chemistry
13.
J Biol Chem ; 292(12): 4942-4952, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28159843

ABSTRACT

Reversible tyrosine phosphorylation is a widespread post-translational modification mechanism underlying cell physiology. Thus, understanding the mechanisms responsible for substrate selection by kinases and phosphatases is central to our ability to model signal transduction at a system level. Classical protein-tyrosine phosphatases can exhibit substrate specificity in vivo by combining intrinsic enzymatic specificity with the network of protein-protein interactions, which positions the enzymes in close proximity to their substrates. Here we use a high throughput approach, based on high density phosphopeptide chips, to determine the in vitro substrate preference of 16 members of the protein-tyrosine phosphatase family. This approach helped identify one residue in the substrate binding pocket of the phosphatase domain that confers specificity for phosphopeptides in a specific sequence context. We also present a Bayesian model that combines intrinsic enzymatic specificity and interaction information in the context of the human protein interaction network to infer new phosphatase substrates at the proteome level.


Subject(s)
Phosphopeptides/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Bayes Theorem , Binding Sites , Humans , Models, Biological , Molecular Docking Simulation , Phosphopeptides/chemistry , Phosphorylation , Protein Conformation , Protein Domains , Protein Interaction Maps , Protein Tyrosine Phosphatases/chemistry , Substrate Specificity
14.
Bioinformatics ; 33(10): 1479-1487, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28073761

ABSTRACT

MOTIVATION: Genome sequencing is producing an ever-increasing amount of associated protein sequences. Few of these sequences have experimentally validated annotations, however, and computational predictions are becoming increasingly successful in producing such annotations. One key challenge remains the prediction of the amino acids in a given protein sequence that are involved in protein-protein interactions. Such predictions are typically based on machine learning methods that take advantage of the properties and sequence positions of amino acids that are known to be involved in interaction. In this paper, we evaluate the importance of various features using Random Forest (RF), and include as a novel feature backbone flexibility predicted from sequences to further optimise protein interface prediction. RESULTS: We observe that there is no single sequence feature that enables pinpointing interacting sites in our Random Forest models. However, combining different properties does increase the performance of interface prediction. Our homomeric-trained RF interface predictor is able to distinguish interface from non-interface residues with an area under the ROC curve of 0.72 in a homomeric test-set. The heteromeric-trained RF interface predictor performs better than existing predictors on a independent heteromeric test-set. We trained a more general predictor on the combined homomeric and heteromeric dataset, and show that in addition to predicting homomeric interfaces, it is also able to pinpoint interface residues in heterodimers. This suggests that our random forest model and the features included capture common properties of both homodimer and heterodimer interfaces. AVAILABILITY AND IMPLEMENTATION: The predictors and test datasets used in our analyses are freely available ( http://www.ibi.vu.nl/downloads/RF_PPI/ ). CONTACT: k.a.feenstra@vu.nl. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Models, Statistical , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Protein Multimerization , Computational Biology/methods , ROC Curve , Sequence Analysis, Protein/methods
15.
Nucleic Acids Res ; 44(8): e72, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26721389

ABSTRACT

Eukaryotic gene expression is regulated by transcription factors (TFs) binding to promoter as well as distal enhancers. TFs recognize short, but specific binding sites (TFBSs) that are located within the promoter and enhancer regions. Functionally relevant TFBSs are often highly conserved during evolution leaving a strong phylogenetic signal. While multiple sequence alignment (MSA) is a potent tool to detect the phylogenetic signal, the current MSA implementations are optimized to align the maximum number of identical nucleotides. This approach might result in the omission of conserved motifs that contain interchangeable nucleotides such as the ETS motif (IUPAC code: GGAW). Here, we introduce ConBind, a novel method to enhance alignment of short motifs, even if their mutual sequence similarity is only partial. ConBind improves the identification of conserved TFBSs by improving the alignment accuracy of TFBS families within orthologous DNA sequences. Functional validation of the Gfi1b + 13 enhancer reveals that ConBind identifies additional functionally important ETS binding sites that were missed by all other tested alignment tools. In addition to the analysis of known regulatory regions, our web tool is useful for the analysis of TFBSs on so far unknown DNA regions identified through ChIP-sequencing.


Subject(s)
Computational Biology/methods , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Sequence Alignment/methods , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Gene Expression Regulation/genetics , Humans , Sequence Analysis, DNA
16.
Bioinformatics ; 32(12): i60-i69, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27307645

ABSTRACT

MOTIVATION: Biological pathways play a key role in most cellular functions. To better understand these functions, diverse computational and cell biology researchers use biological pathway data for various analysis and modeling purposes. For specifying these biological pathways, a community of researchers has defined BioPAX and provided various tools for creating, validating and visualizing BioPAX models. However, a generic software framework for simulating BioPAX models is missing. Here, we attempt to fill this gap by introducing a generic simulation framework for BioPAX. The framework explicitly separates the execution model from the model structure as provided by BioPAX, with the advantage that the modelling process becomes more reproducible and intrinsically more modular; this ensures natural biological constraints are satisfied upon execution. The framework is based on the principles of discrete event systems and multi-agent systems, and is capable of automatically generating a hierarchical multi-agent system for a given BioPAX model. RESULTS: To demonstrate the applicability of the framework, we simulated two types of biological network models: a gene regulatory network modeling the haematopoietic stem cell regulators and a signal transduction network modeling the Wnt/ß-catenin signaling pathway. We observed that the results of the simulations performed using our framework were entirely consistent with the simulation results reported by the researchers who developed the original models in a proprietary language. AVAILABILITY AND IMPLEMENTATION: The framework, implemented in Java, is open source and its source code, documentation and tutorial are available at http://www.ibi.vu.nl/programs/BioASF CONTACT: j.heringa@vu.nl.


Subject(s)
Gene Regulatory Networks , Models, Biological , Signal Transduction , Software , Computer Simulation , Humans , Programming Languages
17.
Bioinformatics ; 32(11): 1678-85, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26342232

ABSTRACT

MOTIVATION: The human microbiome plays a key role in health and disease. Thanks to comparative metatranscriptomics, the cellular functions that are deregulated by the microbiome in disease can now be computationally explored. Unlike gene-centric approaches, pathway-based methods provide a systemic view of such functions; however, they typically consider each pathway in isolation and in its entirety. They can therefore overlook the key differences that (i) span multiple pathways, (ii) contain bidirectionally deregulated components, (iii) are confined to a pathway region. To capture these properties, computational methods that reach beyond the scope of predefined pathways are needed. RESULTS: By integrating an existing module discovery algorithm into comparative metatranscriptomic analysis, we developed metaModules, a novel computational framework for automated identification of the key functional differences between health- and disease-associated communities. Using this framework, we recovered significantly deregulated subnetworks that were indeed recognized to be involved in two well-studied, microbiome-mediated oral diseases, such as butanoate production in periodontal disease and metabolism of sugar alcohols in dental caries. More importantly, our results indicate that our method can be used for hypothesis generation based on automated discovery of novel, disease-related functional subnetworks, which would otherwise require extensive and laborious manual assessment. AVAILABILITY AND IMPLEMENTATION: metaModules is available at https://bitbucket.org/alimay/metamodules/ CONTACT: a.may@vu.nl or s.abeln@vu.nl SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Microbiota , Algorithms , Dental Caries , Humans
18.
Nucleic Acids Res ; 43(W1): W301-5, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25878034

ABSTRACT

Massively parallel sequencing of microbial genetic markers (MGMs) is used to uncover the species composition in a multitude of ecological niches. These sequencing runs often contain a sample with known composition that can be used to evaluate the sequencing quality or to detect novel sequence variants. With NGS-eval, the reads from such (mock) samples can be used to (i) explore the differences between the reads and their references and to (ii) estimate the sequencing error rate. This tool maps these reads to references and calculates as well as visualizes the different types of sequencing errors. Clearly, sequencing errors can only be accurately calculated if the reference sequences are correct. However, even with known strains, it is not straightforward to select the correct references from databases. We previously analysed a pyrosequencing dataset from a mock sample to estimate sequencing error rates and detected sequence variants in our mock community, allowing us to obtain an accurate error estimation. Here, we demonstrate the variant detection and error analysis capability of NGS-eval with Illumina MiSeq reads from the same mock community. While tailored towards the field of metagenomics, this server can be used for any type of MGM-based reads. NGS-eval is available at http://www.ibi.vu.nl/programs/ngsevalwww/.


Subject(s)
Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Metagenomics/methods , Software , Genetic Markers , Internet
19.
Bioinformatics ; 31(19): 3147-55, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26023104

ABSTRACT

MOTIVATION: Integrative network analysis methods provide robust interpretations of differential high-throughput molecular profile measurements. They are often used in a biomedical context-to generate novel hypotheses about the underlying cellular processes or to derive biomarkers for classification and subtyping. The underlying molecular profiles are frequently measured and validated on animal or cellular models. Therefore the results are not immediately transferable to human. In particular, this is also the case in a study of the recently discovered interleukin-17 producing helper T cells (Th17), which are fundamental for anti-microbial immunity but also known to contribute to autoimmune diseases. RESULTS: We propose a mathematical model for finding active subnetwork modules that are conserved between two species. These are sets of genes, one for each species, which (i) induce a connected subnetwork in a species-specific interaction network, (ii) show overall differential behavior and (iii) contain a large number of orthologous genes. We propose a flexible notion of conservation, which turns out to be crucial for the quality of the resulting modules in terms of biological interpretability. We propose an algorithm that finds provably optimal or near-optimal conserved active modules in our model. We apply our algorithm to understand the mechanisms underlying Th17 T cell differentiation in both mouse and human. As a main biological result, we find that the key regulation of Th17 differentiation is conserved between human and mouse. AVAILABILITY AND IMPLEMENTATION: xHeinz, an implementation of our algorithm, as well as all input data and results, are available at http://software.cwi.nl/xheinz and as a Galaxy service at http://services.cbib.u-bordeaux2.fr/galaxy in CBiB Tools. CONTACT: gunnar.klau@cwi.nl SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Data Mining , Models, Biological , Protein Interaction Maps , Animals , Gene Expression Profiling , Humans , Infant, Newborn , Mice , Species Specificity , Th17 Cells/cytology
20.
BMC Bioinformatics ; 16: 325, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26449222

ABSTRACT

BACKGROUND: Protein families participating in protein-protein interactions may contain sub-families that have different binding characteristics, ranging from right binding to showing no interaction at all. Composition differences at the sequence level in these sub-families are often decisive to their differential functional interaction. Methods to predict interface sites from protein sequences typically exploit conservation as a signal. Here, instead, we provide proof of concept that the sequence specificity between interacting versus non-interacting groups can be exploited to recognise interaction sites. RESULTS: We collected homodimeric and monomeric proteins and formed homologous groups, each having an interacting (homodimer) subgroup and a non-interacting (monomer) subgroup. We then compiled multiple sequence alignments of the proteins in the homologous groups and identified compositional differences between the homodimeric and monomeric subgroups for each of the alignment positions. Our results show that this specificity signal distinguishes interface and other surface residues with 40.9% recall and up to 25.1% precision. CONCLUSIONS: To our best knowledge, this is the first large scale study that exploits sequence specificity between interacting and non-interacting homologs to predict interaction sites from sequence information only. The performance obtained indicates that this signal contains valuable information to identify protein-protein interaction sites.


Subject(s)
Proteins/chemistry , Area Under Curve , Dimerization , Protein Interaction Domains and Motifs , Proteins/metabolism , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL