106 research outputs found

    An analysis of the Sargasso Sea resource and the consequences for database composition

    Get PDF
    Background: The environmental sequencing of the Sargasso Sea has introduced a huge new resource of genomic information. Unlike the protein sequences held in the current searchable databases, the Sargasso Sea sequences originate from a single marine environment and have been sequenced from species that are not easily obtainable by laboratory cultivation. The resource also contains very many fragments of whole protein sequences, a side effect of the shotgun sequencing method.These sequences form a significant addendum to the current searchable databases but also present us with some intrinsic difficulties. While it is important to know whether it is possible to assign function to these sequences with the current methods and whether they will increase our capacity to explore sequence space, it is also interesting to know how current bioinformatics techniques will deal with the new sequences in the resource.Results: The Sargasso Sea sequences seem to introduce a bias that decreases the potential of current methods to propose structure and function for new proteins. In particular the high proportion of sequence fragments in the resource seems to result in poor quality multiple alignments.Conclusion: These observations suggest that the new sequences should be used with care, especially if the information is to be used in large scale analyses. On a positive note, the results may just spark improvements in computational and experimental methods to take into account the fragments generated by environmental sequencing techniques

    DISOPRED3: Precise disordered region predictions with annotated protein binding activity

    Get PDF
    Motivation: A sizeable fraction of eukaryotic proteins contain intrinsically disordered regions (IDRs), which act in unfolded states or by undergoing transitions between structured and unstructured conformations. Over time, sequence-based classifiers of IDRs have become fairly accurate and currently a major challenge is linking IDRs to their biological roles from the molecular to the systems level. Results: We describe DISOPRED3, which extends its predecessor with new modules to predict IDRs and protein binding sites within them. Based on recent CASP evaluation results, DISOPRED3 can be regarded as state of the art in the identification of IDRs, and our self-assessment shows that it significantly improves over DISOPRED2 because its predictions are more specific across the whole board and more sensitive to IDRs longer than 20 amino acids. Predicted IDRs are annotated as protein binding through a novel SVM-based classifier, which uses profile data and additional sequence-derived features. Based on benchmarking experiments with full cross-validation, we show that this predictor generates precise assignments of disordered protein binding regions and that it compares well with other publicly available tools. Availability: http://bioinf.cs.ucl.ac.uk/disopred

    Computational Methods for Annotation Transfers from Sequence

    Get PDF
    Surveys of public sequence resources show that experimentally supported functional information is still completely missing for a considerable fraction of known proteins and is clearly incomplete for an even larger portion. Bioinformatics methods have long made use of very diverse data sources alone or in combination to predict protein function, with the understanding that different data types help elucidate complementary biological roles. This chapter focuses on methods accepting amino acid sequences as input and producing GO term assignments directly as outputs; the relevant biological and computational concepts are presented along with the advantages and limitations of individual approaches

    Predicting human protein function with multitask deep neural networks

    Get PDF
    Machine learning methods for protein function prediction are urgently needed, especially now that a substantial fraction of known sequences remains unannotated despite the extensive use of functional assignments based on sequence similarity. One major bottleneck supervised learning faces in protein function prediction is the structured, multi-label nature of the problem, because biological roles are represented by lists of terms from hierarchically organised controlled vocabularies such as the Gene Ontology. In this work, we build on recent developments in the area of deep learning and investigate the usefulness of multitask deep neural networks (MTDNN), which consist of upstream shared layers upon which are stacked in parallel as many independent modules (additional hidden layers with their own output units) as the number of output GO terms (the tasks). MTDNN learns individual tasks partially using shared representations and partially from task-specific characteristics. When no close homologues with experimentally validated functions can be identified, MTDNN gives more accurate predictions than baseline methods based on annotation frequencies in public databases or homology transfers. More importantly, the results show that MTDNN binary classification accuracy is higher than alternative machine learning-based methods that do not exploit commonalities and differences among prediction tasks. Interestingly, compared with a single-task predictor, the performance improvement is not linearly correlated with the number of tasks in MTDNN, but medium size models provide more improvement in our case. One of advantages of MTDNN is that given a set of features, there is no requirement for MTDNN to have a bootstrap feature selection procedure as what traditional machine learning algorithms do. Overall, the results indicate that the proposed MTDNN algorithm improves the performance of protein function prediction. On the other hand, there is still large room for deep learning techniques to further enhance prediction ability

    FFPred 3: feature-based function prediction for all Gene Ontology domains

    Get PDF
    Predicting protein function has been a major goal of bioinformatics for several decades, and it has gained fresh momentum thanks to recent community-wide blind tests aimed at benchmarking available tools on a genomic scale. Sequence-based predictors, especially those performing homology-based transfers, remain the most popular but increasing understanding of their limitations has stimulated the development of complementary approaches, which mostly exploit machine learning. Here we present FFPred 3, which is intended for assigning Gene Ontology terms to human protein chains, when homology with characterized proteins can provide little aid. Predictions are made by scanning the input sequences against an array of Support Vector Machines (SVMs), each examining the relationship between protein function and biophysical attributes describing secondary structure, transmembrane helices, intrinsically disordered regions, signal peptides and other motifs. This update features a larger SVM library that extends its coverage to the cellular component sub-ontology for the first time, prompted by the establishment of a dedicated evaluation category within the Critical Assessment of Functional Annotation. The effectiveness of this approach is demonstrated through benchmarking experiments, and its usefulness is illustrated by analysing the potential functional consequences of alternative splicing in human and their relationship to patterns of biological features

    Evaluation of predictions in the CASP10 model refinement category.

    Get PDF
    Here we report on the assessment results of the third experiment to evaluate the state-of-the-art in protein model refinement, where participants were invited to improve the accuracy of initial protein models for twenty-seven targets. Using an array of complementary evaluation measures, we find that five groups performed better than the naïve (null) method - a marked improvement over CASP9, although only three were significantly better. The leading groups also demonstrated the ability to consistently improve both backbone and side-chain positioning, while other groups reliably enhanced other aspects of protein physicality. The top-ranked group succeeded in improving the backbone in almost 90% of targets, suggesting a strategy that for the first time in CASP refinement is successful in a clear majority of cases. A number of issues remain unsolved: the majority of groups still fail to improve the quality of the starting models; even successful groups were only able to make modest improvements; and no prediction was more similar to the native structure than to the starting model. Successful refinement attempts also often go unrecognized, as suggested by the relatively larger improvements when predictions not submitted as model 1 are also considered. © Proteins 2013;. © 2013 Wiley Periodicals, Inc

    The assessment of methods for protein structure prediction

    Get PDF
    Methods for protein structure prediction are flourishing and becoming widely available to both experimentalists and computational biologists. But, how good are they? What is their range of applicability and how can we know which method is better suited for the task at hand? These are the questions that this chapter tries to address, by describing automatic evaluation methods as well as the world-wide Critical Assessment of Techniques for Protein Structure Prediction (CASP) initiative and focusing on the specific problems of assessing the quality of a protein 3D model

    Using Deep Maxout Neural Networks to improve the accuracy of function prediction from Protein Interaction Networks

    Get PDF
    Protein-protein interaction network data provides valuable information that infers direct links between genes and their biological roles. This information brings a fundamental hypothesis for protein function prediction that interacting proteins tend to have similar functions. With the help of recently-developed network embedding feature generation methods and deep maxout neural networks, it is possible to extract functional representations that encode direct links between protein-protein interactions information and protein function. Our novel method, STRING2GO, successfully adopts deep maxout neural networks to learn functional representations simultaneously encoding both protein-protein interactions and functional predictive information. The experimental results show that STRING2GO outperforms other protein-protein interaction network-based prediction methods and one benchmark method adopted in a recent large scale protein function prediction competition

    The CAFA challenge reports improved protein function prediction and new functional annotations for hundreds of genes through experimental screens

    Get PDF
    The Critical Assessment of Functional Annotation (CAFA) is an ongoing, global, community-driven effort to evaluate and improve the computational annotation of protein function. Here we report on the results of the third CAFA challenge, CAFA3, that featured an expanded analysis over the previous CAFA rounds, both in terms of volume of data analyzed and the types of analysis performed. In a novel and major new development, computational predictions and assessment goals drove some of the experimental assays, resulting in new functional annotations for more than 1000 genes. Specifically, we performed experimental whole-genome mutation screening in Candida albicans and Pseudomonas aureginosa genomes, which provided us with genome-wide experimental data for genes associated with biofilm formation and motility P. aureginosa only). We further performed targeted assays on selected genes in Drosophila melanogaster, which we suspected of being involved in long-term memory. We conclude that, while predictions of the molecular function and biological process annotations have slightly improved over time, those of the cellular component have not. Term-centric prediction of experimental annotations remains equally challenging; although the performance of the top methods is significantly better than expectations set by baseline methods in C. albicans and D. melanogaster, it leaves considerable room and need for improvement. We finally report that the CAFA community now involves a broad range of participants with expertise in bioinformatics, biological experimentation, biocuration, and bio-ontologies, working together to improve functional annotation, computational functio
    • …
    corecore