34 research outputs found

    The development of PIPA: an integrated and automated pipeline for genome-wide protein function annotation

    Get PDF
    BACKGROUND: Automated protein function prediction methods are needed to keep pace with high-throughput sequencing. With the existence of many programs and databases for inferring different protein functions, a pipeline that properly integrates these resources will benefit from the advantages of each method. However, integrated systems usually do not provide mechanisms to generate customized databases to predict particular protein functions. Here, we describe a tool termed PIPA (Pipeline for Protein Annotation) that has these capabilities. RESULTS: PIPA annotates protein functions by combining the results of multiple programs and databases, such as InterPro and the Conserved Domains Database, into common Gene Ontology (GO) terms. The major algorithms implemented in PIPA are: (1) a profile database generation algorithm, which generates customized profile databases to predict particular protein functions, (2) an automated ontology mapping generation algorithm, which maps various classification schemes into GO, and (3) a consensus algorithm to reconcile annotations from the integrated programs and databases. PIPA's profile generation algorithm is employed to construct the enzyme profile database CatFam, which predicts catalytic functions described by Enzyme Commission (EC) numbers. Validation tests show that CatFam yields average recall and precision larger than 95.0%. CatFam is integrated with PIPA. We use an association rule mining algorithm to automatically generate mappings between terms of two ontologies from annotated sample proteins. Incorporating the ontologies' hierarchical topology into the algorithm increases the number of generated mappings. In particular, it generates 40.0% additional mappings from the Clusters of Orthologous Groups (COG) to EC numbers and a six-fold increase in mappings from COG to GO terms. The mappings to EC numbers show a very high precision (99.8%) and recall (96.6%), while the mappings to GO terms show moderate precision (80.0%) and low recall (33.0%). Our consensus algorithm for GO annotation is based on the computation and propagation of likelihood scores associated with GO terms. The test results suggest that, for a given recall, the application of the consensus algorithm yields higher precision than when consensus is not used. CONCLUSION: The algorithms implemented in PIPA provide automated genome-wide protein function annotation based on reconciled predictions from multiple resources

    PSPP: A Protein Structure Prediction Pipeline for Computing Clusters

    Get PDF
    BACKGROUND:Protein structures are critical for understanding the mechanisms of biological systems and, subsequently, for drug and vaccine design. Unfortunately, protein sequence data exceed structural data by a factor of more than 200 to 1. This gap can be partially filled by using computational protein structure prediction. While structure prediction Web servers are a notable option, they often restrict the number of sequence queries and/or provide a limited set of prediction methodologies. Therefore, we present a standalone protein structure prediction software package suitable for high-throughput structural genomic applications that performs all three classes of prediction methodologies: comparative modeling, fold recognition, and ab initio. This software can be deployed on a user's own high-performance computing cluster. METHODOLOGY/PRINCIPAL FINDINGS:The pipeline consists of a Perl core that integrates more than 20 individual software packages and databases, most of which are freely available from other research laboratories. The query protein sequences are first divided into domains either by domain boundary recognition or Bayesian statistics. The structures of the individual domains are then predicted using template-based modeling or ab initio modeling. The predicted models are scored with a statistical potential and an all-atom force field. The top-scoring ab initio models are annotated by structural comparison against the Structural Classification of Proteins (SCOP) fold database. Furthermore, secondary structure, solvent accessibility, transmembrane helices, and structural disorder are predicted. The results are generated in text, tab-delimited, and hypertext markup language (HTML) formats. So far, the pipeline has been used to study viral and bacterial proteomes. CONCLUSIONS:The standalone pipeline that we introduce here, unlike protein structure prediction Web servers, allows users to devote their own computing assets to process a potentially unlimited number of queries as well as perform resource-intensive ab initio structure prediction

    New Onto-Tools: Promoter-Express, nsSNPCounter and Onto-Translate

    Get PDF
    The Onto-Tools suite is composed of an annotation database and eight complementary, web-accessible data mining tools: Onto-Express, Onto-Compare, Onto-Design, Onto-Translate, Onto-Miner, Pathway-Express, Promoter-Express and nsSNPCounter. Promoter-Express is a new tool added to the Onto-Tools ensemble that facilitates the identification of transcription factor binding sites active in specific conditions. nsSNPCounter is another new tool that allows computation and analysis of synonymous and non-synonymous codon substitutions for studying evolutionary rates of protein coding genes. Onto-Translate has also been enhanced to expand its scope and accuracy by fully utilizing the capabilities of the Onto-Tools database. Currently, Onto-Translate allows arbitrary mappings between 28 types of IDs for 53 organisms. Onto-Tools are freely available at

    QuartetS: a fast and accurate algorithm for large-scale orthology detection

    Get PDF
    The unparalleled growth in the availability of genomic data offers both a challenge to develop orthology detection methods that are simultaneously accurate and high throughput and an opportunity to improve orthology detection by leveraging evolutionary evidence in the accumulated sequenced genomes. Here, we report a novel orthology detection method, termed QuartetS, that exploits evolutionary evidence in a computationally efficient manner. Based on the well-established evolutionary concept that gene duplication events can be used to discriminate homologous genes, QuartetS uses an approximate phylogenetic analysis of quartet gene trees to infer the occurrence of duplication events and discriminate paralogous from orthologous genes. We used function- and phylogeny-based metrics to perform a large-scale, systematic comparison of the orthology predictions of QuartetS with those of four other methods [bi-directional best hit (BBH), outgroup, OMA and QuartetS-C (QuartetS followed by clustering)], involving 624 bacterial genomes and >2 million genes. We found that QuartetS slightly, but consistently, outperformed the highly specific OMA method and that, while consuming only 0.5% additional computational time, QuartetS predicted 50% more orthologs with a 50% lower false positive rate than the widely used BBH method. We conclude that, for large-scale phylogenetic and functional analysis, QuartetS and QuartetS-C should be preferred, respectively, in applications where high accuracy and high throughput are required

    QuartetS-DB: a large-scale orthology database for prokaryotes and eukaryotes inferred by evolutionary evidence

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The concept of orthology is key to decoding evolutionary relationships among genes across different species using comparative genomics. QuartetS is a recently reported algorithm for large-scale orthology detection. Based on the well-established evolutionary principle that gene duplication events discriminate paralogous from orthologous genes, QuartetS has been shown to improve orthology detection accuracy while maintaining computational efficiency.</p> <p>Description</p> <p>QuartetS-DB is a new orthology database constructed using the QuartetS algorithm. The database provides orthology predictions among 1621 complete genomes (1365 bacterial, 92 archaeal, and 164 eukaryotic), covering more than seven million proteins and four million pairwise orthologs. It is a major source of orthologous groups, containing more than 300,000 groups of orthologous proteins and 236,000 corresponding gene trees. The database also provides over 500,000 groups of inparalogs. In addition to its size, a distinguishing feature of QuartetS-DB is the ability to allow users to select a cutoff value that modulates the balance between prediction accuracy and coverage of the retrieved pairwise orthologs. The database is accessible at https://applications.bioanalysis.org/quartetsdb.</p> <p>Conclusions</p> <p>QuartetS-DB is one of the largest orthology resources available to date. Because its orthology predictions are underpinned by evolutionary evidence obtained from sequenced genomes, we expect its accuracy to continue to increase in future releases as the genomes of additional species are sequenced.</p

    vNN Web Server for ADMET Predictions

    No full text
    In drug development, early assessments of pharmacokinetic and toxic properties are important stepping stones to avoid costly and unnecessary failures. Considerable progress has recently been made in the development of computer-based (in silico) models to estimate such properties. Nonetheless, such models can be further improved in terms of their ability to make predictions more rapidly, easily, and with greater reliability. To address this issue, we have used our vNN method to develop 15 absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction models. These models quickly assess some of the most important properties of potential drug candidates, including their cytotoxicity, mutagenicity, cardiotoxicity, drug-drug interactions, microsomal stability, and likelihood of causing drug-induced liver injury. Here we summarize the ability of each of these models to predict such properties and discuss their overall performance. All of these ADMET models are publically available on our website (https://vnnadmet.bhsai.org/), which also offers the capability of using the vNN method to customize and build new models

    A Web-Accessible Protein Structure Prediction Pipeline

    No full text
    Proteins are the molecular basis of nearly all structural, catalytic, sensory, and regulatory functions in living organisms. The biological function of a protein is inextricably linked to its three-dimensional (3D) atomic structure. Traditional structure determination methods, such as X-ray and nuclear magnetic resonance techniques, are time-consuming, expensive, and infeasible for the millions of proteins that have been sequenced so far from various organisms. Alternatively, computational structure prediction methods provide a faster and more cost-effective, albeit approximate, alternative to experimental structure determination. We present a high-throughput protein structure prediction pipeline (dubbed “PSPP”), which given input protein sequences infers their 3D atomic structures. The pipeline was designed to be used with high performance computing clusters and to scale with the number of processors. The pipeline encompasses a core Perl module, a parallel job manager, and a Web browser graphical user interface accessible at our Website (www.bhsai.org). The software is currently installed at the Department of Defense (DoD) Maui High Performance Computing Center, and it is available for download along with its associated databases from our site. Currently, DoD scientists are using the pipeline in basic science and drug and vaccine development projects

    AGeS: a software system for microbial genome sequence annotation.

    Get PDF
    BACKGROUND: The annotation of genomes from next-generation sequencing platforms needs to be rapid, high-throughput, and fully integrated and automated. Although a few Web-based annotation services have recently become available, they may not be the best solution for researchers that need to annotate a large number of genomes, possibly including proprietary data, and store them locally for further analysis. To address this need, we developed a standalone software application, the Annotation of microbial Genome Sequences (AGeS) system, which incorporates publicly available and in-house-developed bioinformatics tools and databases, many of which are parallelized for high-throughput performance. METHODOLOGY: The AGeS system supports three main capabilities. The first is the storage of input contig sequences and the resulting annotation data in a central, customized database. The second is the annotation of microbial genomes using an integrated software pipeline, which first analyzes contigs from high-throughput sequencing by locating genomic regions that code for proteins, RNA, and other genomic elements through the Do-It-Yourself Annotation (DIYA) framework. The identified protein-coding regions are then functionally annotated using the in-house-developed Pipeline for Protein Annotation (PIPA). The third capability is the visualization of annotated sequences using GBrowse. To date, we have implemented these capabilities for bacterial genomes. AGeS was evaluated by comparing its genome annotations with those provided by three other methods. Our results indicate that the software tools integrated into AGeS provide annotations that are in general agreement with those provided by the compared methods. This is demonstrated by a >94% overlap in the number of identified genes, a significant number of identical annotated features, and a >90% agreement in enzyme function predictions

    The development of PIPA: an integrated and automated pipeline for genome-wide protein function annotation-0

    No full text
    <p><b>Copyright information:</b></p><p>Taken from "The development of PIPA: an integrated and automated pipeline for genome-wide protein function annotation"</p><p>http://www.biomedcentral.com/1471-2105/9/52</p><p>BMC Bioinformatics 2008;9():52-52.</p><p>Published online 25 Jan 2008</p><p>PMCID:PMC2259298.</p><p></p>enable user access to and control of the pipeline's parallel execution of multiple programs. The execution module wraps the core module, containing all integrated methods (programs and databases), the terminology conversion program, and the consensus annotation program. The support module contains the profile database generation program, which creates new profile databases, and the GO-mapping generation program, which creates GO mappings for the terminology conversion program
    corecore