555 research outputs found

    Distance-based Protein Folding Powered by Deep Learning

    Full text link
    Contact-assisted protein folding has made very good progress, but two challenges remain. One is accurate contact prediction for proteins lack of many sequence homologs and the other is that time-consuming folding simulation is often needed to predict good 3D models from predicted contacts. We show that protein distance matrix can be predicted well by deep learning and then directly used to construct 3D models without folding simulation at all. Using distance geometry to construct 3D models from our predicted distance matrices, we successfully folded 21 of the 37 CASP12 hard targets with a median family size of 58 effective sequence homologs within 4 hours on a Linux computer of 20 CPUs. In contrast, contacts predicted by direct coupling analysis (DCA) cannot fold any of them in the absence of folding simulation and the best CASP12 group folded 11 of them by integrating predicted contacts into complex, fragment-based folding simulation. The rigorous experimental validation on 15 CASP13 targets show that among the 3 hardest targets of new fold our distance-based folding servers successfully folded 2 large ones with <150 sequence homologs while the other servers failed on all three, and that our ab initio folding server also predicted the best, high-quality 3D model for a large homology modeling target. Further experimental validation in CAMEO shows that our ab initio folding server predicted correct fold for a membrane protein of new fold with 200 residues and 229 sequence homologs while all the other servers failed. These results imply that deep learning offers an efficient and accurate solution for ab initio folding on a personal computer

    Structure-Based Sequence Alignment of the Transmembrane Domains of All Human GPCRs: Phylogenetic, Structural and Functional Implications

    Get PDF
    The understanding of G-protein coupled receptors (GPCRs) is undergoing a revolution due to increased information about their signaling and the experimental determination of structures for more than 25 receptors. The availability of at least one receptor structure for each of the GPCR classes, well separated in sequence space, enables an integrated superfamily-wide analysis to identify signatures involving the role of conserved residues, conserved contacts, and downstream signaling in the context of receptor structures. In this study, we align the transmembrane (TM) domains of all experimental GPCR structures to maximize the conserved inter-helical contacts. The resulting superfamily-wide GpcR Sequence-Structure (GRoSS) alignment of the TM domains for all human GPCR sequences is sufficient to generate a phylogenetic tree that correctly distinguishes all different GPCR classes, suggesting that the class-level differences in the GPCR superfamily are encoded at least partly in the TM domains. The inter-helical contacts conserved across all GPCR classes describe the evolutionarily conserved GPCR structural fold. The corresponding structural alignment of the inactive and active conformations, available for a few GPCRs, identifies activation hot-spot residues in the TM domains that get rewired upon activation. Many GPCR mutations, known to alter receptor signaling and cause disease, are located at these conserved contact and activation hot-spot residue positions. The GRoSS alignment places the chemosensory receptor subfamilies for bitter taste (TAS2R) and pheromones (Vomeronasal, VN1R) in the rhodopsin family, known to contain the chemosensory olfactory receptor subfamily. The GRoSS alignment also enables the quantification of the structural variability in the TM regions of experimental structures, useful for homology modeling and structure prediction of receptors. Furthermore, this alignment identifies structurally and functionally important residues in all human GPCRs. These residues can be used to make testable hypotheses about the structural basis of receptor function and about the molecular basis of disease-associated single nucleotide polymorphisms

    Information Theory in Molecular Evolution: From Models to Structures and Dynamics

    Get PDF
    This Special Issue collects novel contributions from scientists in the interdisciplinary field of biomolecular evolution. Works listed here use information theoretical concepts as a core but are tightly integrated with the study of molecular processes. Applications include the analysis of phylogenetic signals to elucidate biomolecular structure and function, the study and quantification of structural dynamics and allostery, as well as models of molecular interaction specificity inspired by evolutionary cues

    Comparative Genomics of Microbial Chemoreceptor Sequence, Structure, and Function

    Get PDF
    Microbial chemotaxis receptors (chemoreceptors) are complex proteins that sense the external environment and signal for flagella-mediated motility, serving as the GPS of the cell. In order to sense a myriad of physicochemical signals and adapt to diverse environmental niches, sensory regions of chemoreceptors are frenetically duplicated, mutated, or lost. Conversely, the chemoreceptor signaling region is a highly conserved protein domain. Extreme conservation of this domain is necessary because it determines very specific helical secondary, tertiary, and quaternary structures of the protein while simultaneously choreographing a network of interactions with the adaptor protein CheW and the histidine kinase CheA. This dichotomous nature has split the chemoreceptor community into two major camps, studying either an organism’s sensory capabilities and physiology or the molecular signal transduction mechanism. Fortunately, the current vast wealth of sequencing data has enabled comparative study of chemoreceptors. Comparative genomics can serve as a bridge between these communities, connecting sequence, structure, and function through comprehensive studies on scales ranging from minute and molecular to global and ecological. Herein are four works in which comparative genomics illuminates unanswered questions across the broad chemoreceptor landscape. First, we used evolutionary histories to refine chemoreceptor interactions in Thermotoga maritima, pairing phylogenetics with x-ray crystallography. Next, we uncovered the origin of a unique chemoreceptor, isolated only from hypervirulent strains of Campylobacter jejuni, by comparing chemoreceptor signaling and sensory regions from Campylobacter and Helicobacter. We then selected the opportunistic human pathogen Pseudomonas aeruginosa to address the question of assigning multiple chemoreceptors to multiple chemotaxis pathways within the same organism. We assigned all P. aeruginosa receptors to pathways using a novel in silico approach by incorporating sequence information spanning the entire taxonomic order Pseudomonadales and beyond. Finally, we surveyed the chemotaxis systems of all environmental, commensal, laboratory, and pathogenic strains of the ubiquitous Escherichia coli, where we discovered an ancestral chemoreceptor gene loss event that may have predisposed a well-studied subpopulation to adopt extra-intestinal pathogenic lifestyles. Overall, comparative genomics is a cutting edge method for comprehensive chemoreceptor study that is poised to promote synergy within and expand the significance of the chemoreceptor field

    Mass & secondary structure propensity of amino acids explain their mutability and evolutionary replacements

    Get PDF
    Why is an amino acid replacement in a protein accepted during evolution? The answer given by bioinformatics relies on the frequency of change of each amino acid by another one and the propensity of each to remain unchanged. We propose that these replacement rules are recoverable from the secondary structural trends of amino acids. A distance measure between high-resolution Ramachandran distributions reveals that structurally similar residues coincide with those found in substitution matrices such as BLOSUM: Asn Asp, Phe Tyr, Lys Arg, Gln Glu, Ile Val, Met → Leu; with Ala, Cys, His, Gly, Ser, Pro, and Thr, as structurally idiosyncratic residues. We also found a high average correlation (\overline{R} R = 0.85) between thirty amino acid mutability scales and the mutational inertia (I X ), which measures the energetic cost weighted by the number of observations at the most probable amino acid conformation. These results indicate that amino acid substitutions follow two optimally-efficient principles: (a) amino acids interchangeability privileges their secondary structural similarity, and (b) the amino acid mutability depends directly on its biosynthetic energy cost, and inversely with its frequency. These two principles are the underlying rules governing the observed amino acid substitutions. © 2017 The Author(s)

    Homology modeling in the time of collective and artificial intelligence

    Get PDF
    Homology modeling is a method for building protein 3D structures using protein primary sequence and utilizing prior knowledge gained from structural similarities with other proteins. The homology modeling process is done in sequential steps where sequence/structure alignment is optimized, then a backbone is built and later, side-chains are added. Once the low-homology loops are modeled, the whole 3D structure is optimized and validated. In the past three decades, a few collective and collaborative initiatives allowed for continuous progress in both homology and ab initio modeling. Critical Assessment of protein Structure Prediction (CASP) is a worldwide community experiment that has historically recorded the progress in this field. Folding@Home and Rosetta@Home are examples of crowd-sourcing initiatives where the community is sharing computational resources, whereas RosettaCommons is an example of an initiative where a community is sharing a codebase for the development of computational algorithms. Foldit is another initiative where participants compete with each other in a protein folding video game to predict 3D structure. In the past few years, contact maps deep machine learning was introduced to the 3D structure prediction process, adding more information and increasing the accuracy of models significantly. In this review, we will take the reader in a journey of exploration from the beginnings to the most recent turnabouts, which have revolutionized the field of homology modeling. Moreover, we discuss the new trends emerging in this rapidly growing field.O

    GenSeed-HMM: A tool for progressive assembly using profile HMMs as seeds and its application in Alpavirinae viral discovery from metagenomic data

    Get PDF
    This work reports the development of GenSeed-HMM, a program that implements seed-driven progressive assembly, an approach to reconstruct specific sequences from unassembled data, starting from short nucleotide or protein seed sequences or profile Hidden Markov Models (HMM). The program can use any one of a number of sequence assemblers. Assembly is performed in multiple steps and relatively few reads are used in each cycle, consequently the program demands low computational resources. As a proof-of-concept and to demonstrate the power of HMM-driven progressive assemblies, GenSeed-HMM was applied to metagenomic datasets in the search for diverse ssDNA bacteriophages from the recently described Alpavirinae subfamily. Profile HMMs were built using Alpavirinae-specific regions from multiple sequence alignments using either the viral protein 1 (VP1) (major capsid protein) or VP4 (genome replication initiation protein). These profile HMMs were used by GenSeed-HMM (running Newbler assembler) as seeds to reconstruct viral genomes from sequencing datasets of human fecal samples. All contigs obtained were annotated and taxonomically classified using similarity searches and phylogenetic analyses. The most specific profile HMM seed enabled the reconstruction of 45 partial or complete Alpavirinae genomic sequences. A comparison with conventional (global) assembly of the same original dataset, using Newbler in a standalone execution, revealed that GenSeed-HMM outperformed global genomic assembly in several metrics employed. This approach is capable of detecting organisms that have not been used in the construction of the profile HMM, which opens up the possibility of diagnosing novel viruses, without previous specific information, constituting a de novo diagnosis. Additional applications include, but are not limited to, the specific assembly of extrachromosomal elements such as plastid and mitochondrial genomes from metagenomic data. Profile HMM seeds can also be used to reconstruct specific protein coding genes for gene diversity studies, and to determine all possible gene variants present in a metagenomic sample. Such surveys could be useful to detect the emergence of drug-resistance variants in sensitive environments such as hospitals and animal production facilities, where antibiotics are regularly used. Finally, GenSeed-HMM can be used as an adjunct for gap closure on assembly finishing projects, by using multiple contig ends as anchored seeds

    Enhancing Evolutionary Couplings with Deep Convolutional Neural Networks

    Get PDF
    While genes are defined by sequence, in biological systems a protein's function is largely determined by its three-dimensional structure. Evolutionary information embedded within multiple sequence alignments provides a rich source of data for inferring structural constraints on macromolecules. Still, many proteins of interest lack sufficient numbers of related sequences, leading to noisy, error-prone residue-residue contact predictions. Here we introduce DeepContact, a convolutional neural network (CNN)-based approach that discovers co-evolutionary motifs and leverages these patterns to enable accurate inference of contact probabilities, particularly when few related sequences are available. DeepContact significantly improves performance over previous methods, including in the CASP12 blind contact prediction task where we achieved top performance with another CNN-based approach. Moreover, our tool converts hard-to-interpret coupling scores into probabilities, moving the field toward a consistent metric to assess contact prediction across diverse proteins. Through substantially improving the precision-recall behavior of contact prediction, DeepContact suggests we are near a paradigm shift in template-free modeling for protein structure prediction. Many protein structures of interest remain out of reach for both computational prediction and experimental determination. DeepContact learns patterns of co-evolution across thousands of experimentally determined structures, identifying conserved local motifs and leveraging this information to improve protein residue-residue contact predictions. DeepContact extracts additional information from the evolutionary couplings using its knowledge of co-evolution and structural space, while also converting coupling scores into probabilities that are comparable across protein sequences and alignments. Keywords: contact prediction; convolutional neural networks; deep learning; protein structure prediction; structure prediction; co-evolution; evolutionary couplingsNational Institutes of Health (U.S.) (Grant R01GM081871
    • …
    corecore