Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Comput Math Methods Med ; 2022: 7191684, 2022.
Article in English | MEDLINE | ID: mdl-35242211

ABSTRACT

Protein-protein interactions (PPIs) play a crucial role in understanding disease pathogenesis, genetic mechanisms, guiding drug design, and other biochemical processes, thus, the identification of PPIs is of great importance. With the rapid development of high-throughput sequencing technology, a large amount of PPIs sequence data has been accumulated. Researchers have designed many experimental methods to detect PPIs by using these sequence data, hence, the prediction of PPIs has become a research hotspot in proteomics. However, since traditional experimental methods are both time-consuming and costly, it is difficult to analyze and predict the massive amount of PPI data quickly and accurately. To address these issues, many computational systems employing machine learning knowledge were widely applied to PPIs prediction, thereby improving the overall recognition rate. In this paper, a novel and efficient computational technology is presented to implement a protein interaction prediction system using only protein sequence information. First, the Position-Specific Iterated Basic Local Alignment Search Tool (PSI-BLAST) was employed to generate a position-specific scoring matrix (PSSM) containing protein evolutionary information from the initial protein sequence. Second, we used a novel data processing feature representation scheme, MatFLDA, to extract the essential information of PSSM for protein sequences and obtained five training and five testing datasets by adopting a five-fold cross-validation method. Finally, the random fern (RFs) classifier was employed to infer the interactions among proteins, and a model called MatFLDA_RFs was developed. The proposed MatFLDA_RFs model achieved good prediction performance with 95.03% average accuracy on Yeast dataset and 85.35% average accuracy on H. pylori dataset, which effectively outperformed other existing computational methods. The experimental results indicate that the proposed method is capable of yielding better prediction results of PPIs, which provides an effective tool for the detection of new PPIs and the in-depth study of proteomics. Finally, we also developed a web server for the proposed model to predict protein-protein interactions, which is freely accessible online at http://120.77.11.78:5001/webserver/MatFLDA_RFs.


Subject(s)
Protein Interaction Mapping/methods , Protein Interaction Maps/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Computational Biology , Databases, Protein/statistics & numerical data , Discriminant Analysis , Evolution, Molecular , Helicobacter pylori/genetics , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Machine Learning , Position-Specific Scoring Matrices , Protein Interaction Mapping/statistics & numerical data , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment/methods , Sequence Alignment/statistics & numerical data , Support Vector Machine
2.
PLoS Comput Biol ; 17(8): e1008844, 2021 08.
Article in English | MEDLINE | ID: mdl-34370723

ABSTRACT

Many biological processes are mediated by protein-protein interactions (PPIs). Because protein domains are the building blocks of proteins, PPIs likely rely on domain-domain interactions (DDIs). Several attempts exist to infer DDIs from PPI networks but the produced datasets are heterogeneous and sometimes not accessible, while the PPI interactome data keeps growing. We describe a new computational approach called "PPIDM" (Protein-Protein Interactions Domain Miner) for inferring DDIs using multiple sources of PPIs. The approach is an extension of our previously described "CODAC" (Computational Discovery of Direct Associations using Common neighbors) method for inferring new edges in a tripartite graph. The PPIDM method has been applied to seven widely used PPI resources, using as "Gold-Standard" a set of DDIs extracted from 3D structural databases. Overall, PPIDM has produced a dataset of 84,552 non-redundant DDIs. Statistical significance (p-value) is calculated for each source of PPI and used to classify the PPIDM DDIs in Gold (9,175 DDIs), Silver (24,934 DDIs) and Bronze (50,443 DDIs) categories. Dataset comparison reveals that PPIDM has inferred from the 2017 releases of PPI sources about 46% of the DDIs present in the 2020 release of the 3did database, not counting the DDIs present in the Gold-Standard. The PPIDM dataset contains 10,229 DDIs that are consistent with more than 13,300 PPIs extracted from the IMEx database, and nearly 23,300 DDIs (27.5%) that are consistent with more than 214,000 human PPIs extracted from the STRING database. Examples of newly inferred DDIs covering more than 10 PPIs in the IMEx database are provided. Further exploitation of the PPIDM DDI reservoir includes the inventory of possible partners of a protein of interest and characterization of protein interactions at the domain level in combination with other methods. The result is publicly available at http://ppidm.loria.fr/.


Subject(s)
Protein Interaction Domains and Motifs , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps , Algorithms , Computational Biology , Data Mining/statistics & numerical data , Databases, Protein/statistics & numerical data , Humans , Software
3.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33787847

ABSTRACT

With the increasing volume of high-throughput sequencing data from a variety of omics techniques in the field of plant-pathogen interactions, sorting, retrieving, processing and visualizing biological information have become a great challenge. Within the explosion of data, machine learning offers powerful tools to process these complex omics data by various algorithms, such as Bayesian reasoning, support vector machine and random forest. Here, we introduce the basic frameworks of machine learning in dissecting plant-pathogen interactions and discuss the applications and advances of machine learning in plant-pathogen interactions from molecular to network biology, including the prediction of pathogen effectors, plant disease resistance protein monitoring and the discovery of protein-protein networks. The aim of this review is to provide a summary of advances in plant defense and pathogen infection and to indicate the important developments of machine learning in phytopathology.


Subject(s)
Host-Pathogen Interactions/genetics , Plant Diseases/genetics , Plant Pathology/statistics & numerical data , Plants/genetics , Protein Interaction Mapping/statistics & numerical data , Support Vector Machine , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bayes Theorem , Disease Resistance/genetics , Fungal Proteins/genetics , Fungal Proteins/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , NLR Proteins/genetics , NLR Proteins/immunology , Pathogen-Associated Molecular Pattern Molecules/chemistry , Pathogen-Associated Molecular Pattern Molecules/immunology , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Diseases/virology , Plants/immunology , Plants/microbiology , Plants/virology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Viral Proteins/genetics , Viral Proteins/immunology
4.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33693513

ABSTRACT

Proteins interact with each other to play critical roles in many biological processes in cells. Although promising, laboratory experiments usually suffer from the disadvantages of being time-consuming and labor-intensive. The results obtained are often not robust and considerably uncertain. Due recently to advances in high-throughput technologies, a large amount of proteomics data has been collected and this presents a significant opportunity and also a challenge to develop computational models to predict protein-protein interactions (PPIs) based on these data. In this paper, we present a comprehensive survey of the recent efforts that have been made towards the development of effective computational models for PPI prediction. The survey introduces the algorithms that can be used to learn computational models for predicting PPIs, and it classifies these models into different categories. To understand their relative merits, the paper discusses different validation schemes and metrics to evaluate the prediction performance. Biological databases that are commonly used in different experiments for performance comparison are also described and their use in a series of extensive experiments to compare different prediction models are discussed. Finally, we present some open issues in PPI prediction for future work. We explain how the performance of PPI prediction can be improved if these issues are effectively tackled.


Subject(s)
Computational Biology/methods , Protein Interaction Mapping/methods , Proteins/metabolism , Software , Support Vector Machine , Databases, Genetic , Databases, Protein , Gene Ontology , Humans , Models, Molecular , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping/statistics & numerical data , Proteins/chemistry , Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
5.
Proteins ; 89(6): 639-647, 2021 06.
Article in English | MEDLINE | ID: mdl-33458895

ABSTRACT

Proteins often exert their function by binding to other cellular partners. The hot spots are key residues for protein-protein binding. Their identification may shed light on the impact of disease associated mutations on protein complexes and help design protein-protein interaction inhibitors for therapy. Unfortunately, current machine learning methods to predict hot spots, suffer from limitations caused by gross errors in the data matrices. Here, we present a novel data pre-processing pipeline that overcomes this problem by recovering a low rank matrix with reduced noise using Robust Principal Component Analysis. Application to existing databases shows the predictive power of the method.


Subject(s)
Machine Learning , Principal Component Analysis , Protein Interaction Mapping/statistics & numerical data , Proteins/chemistry , Binding Sites , Computational Biology/methods , Databases, Protein , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Proteins/metabolism , ROC Curve
6.
Nat Commun ; 11(1): 3862, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737291

ABSTRACT

Allostery in proteins influences various biological processes such as regulation of gene transcription and activities of enzymes and cell signaling. Computational approaches for analysis of allosteric coupling provide inexpensive opportunities to predict mutations and to design small-molecule agents to control protein function and cellular activity. We develop a computationally efficient network-based method, Ohm, to identify and characterize allosteric communication networks within proteins. Unlike previously developed simulation-based approaches, Ohm relies solely on the structure of the protein of interest. We use Ohm to map allosteric networks in a dataset composed of 20 proteins experimentally identified to be allosterically regulated. Further, the Ohm allostery prediction for the protein CheY correlates well with NMR CHESCA studies. Our webserver, Ohm.dokhlab.org, automatically determines allosteric network architecture and identifies critical coupled residues within this network.


Subject(s)
Algorithms , Methyl-Accepting Chemotaxis Proteins/chemistry , Protein Interaction Mapping/statistics & numerical data , Software , Allosteric Regulation , Allosteric Site , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins , Humans , Internet , Methyl-Accepting Chemotaxis Proteins/antagonists & inhibitors , Methyl-Accepting Chemotaxis Proteins/metabolism , Molecular Dynamics Simulation , Protein Structure, Secondary
7.
PLoS One ; 15(1): e0227598, 2020.
Article in English | MEDLINE | ID: mdl-31986158

ABSTRACT

Comparative network analysis provides effective computational means for gaining novel insights into the structural and functional compositions of biological networks. In recent years, various methods have been developed for biological network alignment, whose main goal is to identify important similarities and critical differences between networks in terms of their topology and composition. A major impediment to advancing network alignment techniques has been the lack of gold-standard benchmarks that can be used for accurate and comprehensive performance assessment of such algorithms. The original NAPAbench (network alignment performance assessment benchmark) was developed to address this problem, and it has been widely utilized by many researchers for the development, evaluation, and comparison of novel network alignment techniques. In this work, we introduce NAPAbench 2-a major update of the original NAPAbench that was introduced in 2012. NAPAbench 2 includes a completely redesigned network synthesis algorithm that can generate protein-protein interaction (PPI) network families whose characteristics closely match those of the latest real PPI networks. Furthermore, the network synthesis algorithm comes with an intuitive GUI that allows users to easily generate PPI network families with an arbitrary number of networks of any size, according to a flexible user-defined phylogeny. In addition, NAPAbench 2 provides updated benchmark datasets-created using the redesigned network synthesis algorithm-which can be used for comprehensive performance assessment of network alignment algorithms and their scalability.


Subject(s)
Algorithms , Computational Biology/methods , Protein Interaction Mapping/methods , Databases, Factual , Phylogeny , Protein Interaction Mapping/statistics & numerical data
8.
Comput Math Methods Med ; 2019: 5238406, 2019.
Article in English | MEDLINE | ID: mdl-31531123

ABSTRACT

Protein-protein interactions (PPIs) play a crucial role in various biological processes. To better comprehend the pathogenesis and treatments of various diseases, it is necessary to learn the detail of these interactions. However, the current experimental method still has many false-positive and false-negative problems. Computational prediction of protein-protein interaction has become a more important prediction method which can overcome the obstacles of the experimental method. In this work, we proposed a novel computational domain-based method for PPI prediction, and an SVM model for the prediction was built based on the physicochemical property of the domain. The outcomes of SVM and the domain-domain score were used to construct the prediction model for protein-protein interaction. The predicted results demonstrated the domain-based research can enhance the ability to predict protein interactions.


Subject(s)
Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Databases, Protein/statistics & numerical data , Humans , Mathematical Concepts , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps , Proteins/chemistry , Support Vector Machine
9.
PLoS Comput Biol ; 15(8): e1007239, 2019 08.
Article in English | MEDLINE | ID: mdl-31437145

ABSTRACT

Tailored therapy aims to cure cancer patients effectively and safely, based on the complex interactions between patients' genomic features, disease pathology and drug metabolism. Thus, the continual increase in scientific literature drives the need for efficient methods of data mining to improve the extraction of useful information from texts based on patients' genomic features. An important application of text mining to tailored therapy in cancer encompasses the use of mutations and cancer fusion genes as moieties that change patients' cellular networks to develop cancer, and also affect drug metabolism. Fusion proteins, which are derived from the slippage of two parental genes, are produced in cancer by chromosomal aberrations and trans-splicing. Given that the two parental proteins for predicted fusion proteins are known, we used our previously developed method for identifying chimeric protein-protein interactions (ChiPPIs) associated with the fusion proteins. Here, we present a validation approach that receives fusion proteins of interest, predicts their cellular network alterations by ChiPPI and validates them by our new method, ProtFus, using an online literature search. This process resulted in a set of 358 fusion proteins and their corresponding protein interactions, as a training set for a Naïve Bayes classifier, to identify predicted fusion proteins that have reliable evidence in the literature and that were confirmed experimentally. Next, for a test group of 1817 fusion proteins, we were able to identify from the literature 2908 PPIs in total, across 18 cancer types. The described method, ProtFus, can be used for screening the literature to identify unique cases of fusion proteins and their PPIs, as means of studying alterations of protein networks in cancers. Availability: http://protfus.md.biu.ac.il/.


Subject(s)
Data Mining/methods , Oncogene Proteins, Fusion/genetics , Protein Interaction Mapping/methods , Algorithms , Bayes Theorem , Big Data , Computational Biology , Data Mining/statistics & numerical data , Databases, Genetic , Humans , Mutation , Neoplasms/genetics , Neoplasms/therapy , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Precision Medicine , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps
10.
PLoS Comput Biol ; 15(4): e1006888, 2019 04.
Article in English | MEDLINE | ID: mdl-30995217

ABSTRACT

In response to a need for improved treatments, a number of promising novel targeted cancer therapies are being developed that exploit human synthetic lethal interactions. This is facilitating personalised medicine strategies in cancers where specific tumour suppressors have become inactivated. Mainly due to the constraints of the experimental procedures, relatively few human synthetic lethal interactions have been identified. Here we describe SLant (Synthetic Lethal analysis via Network topology), a computational systems approach to predicting human synthetic lethal interactions that works by identifying and exploiting conserved patterns in protein interaction network topology both within and across species. SLant out-performs previous attempts to classify human SSL interactions and experimental validation of the models predictions suggests it may provide useful guidance for future SSL screenings and ultimately aid targeted cancer therapy development.


Subject(s)
Protein Interaction Maps/genetics , Synthetic Lethal Mutations , Algorithms , Animals , Artificial Intelligence , Computational Biology , Drug Discovery , Gene Ontology , Genes, Essential , Humans , Models, Biological , Molecular Targeted Therapy , Multigene Family , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps/drug effects , Synthetic Biology , Synthetic Lethal Mutations/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
J Proteome Res ; 18(5): 2052-2064, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30931570

ABSTRACT

Proteomics data analysis strongly benefits from not studying single proteins in isolation but taking their multivariate interdependence into account. We introduce PerseusNet, the new Perseus network module for the biological analysis of proteomics data. Proteomics is commonly used to generate networks, e.g., with affinity purification experiments, but networks are also used to explore proteomics data. PerseusNet supports the biomedical researcher for both modes of data analysis with a multitude of activities. For affinity purification, a volcano-plot-based statistical analysis method for network generation is featured which is scalable to large numbers of baits. For posttranslational modifications of proteins, such as phosphorylation, a collection of dedicated network analysis tools helps in elucidating cellular signaling events. Co-expression network analysis of proteomics data adopts established tools from transcriptome co-expression analysis. PerseusNet is extensible through a plugin architecture in a multi-lingual way, integrating analyses in C#, Python, and R, and is freely available at http://www.perseus-framework.org .


Subject(s)
Computational Biology/methods , Protein Interaction Mapping/statistics & numerical data , Protein Processing, Post-Translational , Proteome/metabolism , Software , Animals , Computational Biology/statistics & numerical data , Data Interpretation, Statistical , Gene Regulatory Networks , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Proteome/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Nat Commun ; 10(1): 1118, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850613

ABSTRACT

It remains a significant challenge to define individual protein associations within networks where an individual protein can directly interact with other proteins and/or be part of large complexes, which contain functional modules. Here we demonstrate the topological scoring (TopS) algorithm for the analysis of quantitative proteomic datasets from affinity purifications. Data is analyzed in a parallel fashion where a prey protein is scored in an individual affinity purification by aggregating information from the entire dataset. Topological scores span a broad range of values indicating the enrichment of an individual protein in every bait protein purification. TopS is applied to interaction networks derived from human DNA repair proteins and yeast chromatin remodeling complexes. TopS highlights potential direct protein interactions and modules within complexes. TopS is a rapid method for the efficient and informative computational analysis of datasets, is complementary to existing analysis pipelines, and provides important insights into protein interaction networks.


Subject(s)
Algorithms , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps , Chromatin Assembly and Disassembly , DNA Repair , Databases, Protein/statistics & numerical data , Humans , Likelihood Functions , Proteomics/statistics & numerical data , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
13.
J Bioinform Comput Biol ; 17(1): 1950001, 2019 02.
Article in English | MEDLINE | ID: mdl-30803297

ABSTRACT

The prediction of protein complexes based on the protein interaction network is a fundamental task for the understanding of cellular life as well as the mechanisms underlying complex disease. A great number of methods have been developed to predict protein complexes based on protein-protein interaction (PPI) networks in recent years. However, because the high throughput data obtained from experimental biotechnology are incomplete, and usually contain a large number of spurious interactions, most of the network-based protein complex identification methods are sensitive to the reliability of the PPI network. In this paper, we propose a new method, Identification of Protein Complex based on Refined Protein Interaction Network (IPC-RPIN), which integrates the topology, gene expression profiles and GO functional annotation information to predict protein complexes from the reconstructed networks. To demonstrate the performance of the IPC-RPIN method, we evaluated the IPC-RPIN on three PPI networks of Saccharomycescerevisiae and compared it with four state-of-the-art methods. The simulation results show that the IPC-RPIN achieved a better result than the other methods on most of the measurements and is able to discover small protein complexes which have traditionally been neglected.


Subject(s)
Gene Ontology/statistics & numerical data , Protein Interaction Maps , Transcriptome , Algorithms , Cluster Analysis , Computational Biology , Databases, Protein/statistics & numerical data , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Interaction Mapping/statistics & numerical data , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
14.
Brief Bioinform ; 20(1): 274-287, 2019 01 18.
Article in English | MEDLINE | ID: mdl-29028906

ABSTRACT

The identification of plant-pathogen protein-protein interactions (PPIs) is an attractive and challenging research topic for deciphering the complex molecular mechanism of plant immunity and pathogen infection. Considering that the experimental identification of plant-pathogen PPIs is time-consuming and labor-intensive, computational methods are emerging as an important strategy to complement the experimental methods. In this work, we first evaluated the performance of traditional computational methods such as interolog, domain-domain interaction and domain-motif interaction in predicting known plant-pathogen PPIs. Owing to the low sensitivity of the traditional methods, we utilized Random Forest to build an inter-species PPI prediction model based on multiple sequence encodings and novel network attributes in the established plant PPI network. Critical assessment of the features demonstrated that the integration of sequence information and network attributes resulted in significant and robust performance improvement. Additionally, we also discussed the influence of Gene Ontology and gene expression information on the prediction performance. The Web server implementing the integrated prediction method, named InterSPPI, has been made freely available at http://systbio.cau.edu.cn/intersppi/index.php. InterSPPI could achieve a reasonably high accuracy with a precision of 73.8% and a recall of 76.6% in the independent test. To examine the applicability of InterSPPI, we also conducted cross-species and proteome-wide plant-pathogen PPI prediction tests. Taken together, we hope this work can provide a comprehensive understanding of the current status of plant-pathogen PPI predictions, and the proposed InterSPPI can become a useful tool to accelerate the exploration of plant-pathogen interactions.


Subject(s)
Plant Proteins/metabolism , Plants/metabolism , Plants/microbiology , Protein Interaction Mapping/methods , Algorithms , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/microbiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/immunology , Arabidopsis Proteins/metabolism , Computational Biology/methods , Databases, Protein/statistics & numerical data , Gene Expression Profiling/statistics & numerical data , Gene Ontology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Machine Learning , Models, Biological , Plant Diseases/genetics , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Immunity/genetics , Plant Proteins/genetics , Plant Proteins/immunology , Plants/genetics , Protein Interaction Mapping/statistics & numerical data
15.
J Proteome Res ; 17(11): 3740-3748, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30265007

ABSTRACT

Metabolic labeling with heavy water followed by LC-MS is a high throughput approach to study proteostasis in vivo. Advances in mass spectrometry and sample processing have allowed consistent detection of thousands of proteins at multiple time points. However, freely available automated bioinformatics tools to analyze and extract protein decay rate constants are lacking. Here, we describe d2ome-a robust, automated software solution for in vivo protein turnover analysis. d2ome is highly scalable, uses innovative approaches to nonlinear fitting, implements Grubbs' outlier detection and removal, uses weighted-averaging of replicates, applies a data dependent elution time windowing, and uses mass accuracy in peak detection. Here, we discuss the application of d2ome in a comparative study of protein turnover in the livers of normal vs Western diet-fed LDLR-/- mice (mouse model of nonalcoholic fatty liver disease), which contained 256 LC-MS experiments. The study revealed reduced stability of 40S ribosomal protein subunits in the Western diet-fed mice.


Subject(s)
Deuterium Oxide/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Proteome/metabolism , Ribosomal Proteins/metabolism , Software , Animals , Chromatography, Liquid , Deuterium Oxide/chemistry , Diet, Western/adverse effects , Disease Models, Animal , Gene Expression , Half-Life , Isotope Labeling/methods , Liver/chemistry , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Protein Interaction Mapping/statistics & numerical data , Proteolysis , Proteome/chemistry , Proteome/genetics , Proteome/isolation & purification , Proteostasis/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/isolation & purification , Tandem Mass Spectrometry
16.
Methods Enzymol ; 603: 221-235, 2018.
Article in English | MEDLINE | ID: mdl-29673528

ABSTRACT

Although general anesthesia induced by inhaled anesthetics produces definitive phenotypes (e.g., loss of mobility, amnesia, analgesia), the underlying targets of these drugs are still not clear. Genomics and proteomic techniques are discussed for measurement of global transcriptional and translational changes after inhaled anesthetic exposures. The current discussion focuses primarily on the genomic and proteomic technical methodology. We also include a discussion of network and pathway analyses for data interpretation after identification of the targets.


Subject(s)
Anesthetics, Inhalation/pharmacokinetics , Gene Regulatory Networks , Protein Biosynthesis , Protein Interaction Mapping/statistics & numerical data , Proteogenomics/methods , Transcription, Genetic , Anesthesia, General , Anesthetics, Inhalation/pharmacology , Animals , Carbon Radioisotopes , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Fetus , Halothane/pharmacokinetics , Humans , Isoflurane/pharmacokinetics , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Primary Cell Culture , Protein Binding , Proteogenomics/instrumentation , Rats , Real-Time Polymerase Chain Reaction/methods , Sevoflurane/pharmacokinetics , Staining and Labeling/methods
17.
Brief Bioinform ; 19(5): 995-1007, 2018 09 28.
Article in English | MEDLINE | ID: mdl-28369159

ABSTRACT

Various techniques have been developed for identifying the most probable interactants of a protein under a given biological context. In this article, we dissect the effects of the choice of the protein-protein interaction network (PPI) and the manipulation of PPI settings on the network neighborhood of the influenza A virus (IAV) network, as well as hits in genome-wide small interfering RNA screen results for IAV host factors. We investigate the potential of context filtering, which uses text mining evidence linked to PPI edges, as a complement to the edge confidence scores typically provided in PPIs for filtering, for obtaining more biologically relevant network neighborhoods. Here, we estimate the maximum performance of context filtering to isolate a Kyoto Encyclopedia of Genes and Genomes (KEGG) network Ki from a union of KEGG networks and its network neighborhood. The work gives insights on the use of human PPIs in network neighborhood approaches for functional inference.


Subject(s)
Protein Interaction Maps , Algorithms , Computational Biology/methods , Data Mining , Gene Regulatory Networks , Genome-Wide Association Study/statistics & numerical data , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza A virus/physiology , Protein Interaction Mapping/statistics & numerical data , Protein Interaction Maps/genetics , RNA, Small Interfering/genetics
18.
Pac Symp Biocomput ; 23: 92-103, 2018.
Article in English | MEDLINE | ID: mdl-29218872

ABSTRACT

The emergence of drug resistance to traditional chemotherapy and newer targeted therapies in cancer patients is a major clinical challenge. Reactivation of the same or compensatory signaling pathways is a common class of drug resistance mechanisms. Employing drug combinations that inhibit multiple modules of reactivated signaling pathways is a promising strategy to overcome and prevent the onset of drug resistance. However, with thousands of available FDA-approved and investigational compounds, it is infeasible to experimentally screen millions of possible drug combinations with limited resources. Therefore, computational approaches are needed to constrain the search space and prioritize synergistic drug combinations for preclinical studies. In this study, we propose a novel approach for predicting drug combinations through investigating potential effects of drug targets on disease signaling network. We first construct a disease signaling network by integrating gene expression data with disease-associated driver genes. Individual drugs that can partially perturb the disease signaling network are then selected based on a drug-disease network "impact matrix", which is calculated using network diffusion distance from drug targets to signaling network elements. The selected drugs are subsequently clustered into communities (subgroups), which are proposed to share similar mechanisms of action. Finally, drug combinations are ranked according to maximal impact on signaling sub-networks from distinct mechanism-based communities. Our method is advantageous compared to other approaches in that it does not require large amounts drug dose response data, drug-induced "omics" profiles or clinical efficacy data, which are not often readily available. We validate our approach using a BRAF-mutant melanoma signaling network and combinatorial in vitro drug screening data, and report drug combinations with diverse mechanisms of action and opportunities for drug repositioning.


Subject(s)
Drug Therapy, Combination/methods , Signal Transduction/drug effects , Antineoplastic Combined Chemotherapy Protocols , Computational Biology/methods , Drug Combinations , Drug Repositioning , Drug Resistance , Drug Resistance, Neoplasm , Gene Expression Profiling/statistics & numerical data , Humans , Melanoma/drug therapy , Melanoma/genetics , Mutation , Neoplasms/drug therapy , Protein Interaction Mapping/statistics & numerical data , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics
19.
Pac Symp Biocomput ; 23: 111-122, 2018.
Article in English | MEDLINE | ID: mdl-29218874

ABSTRACT

Discovering disease pathways, which can be defined as sets of proteins associated with a given disease, is an important problem that has the potential to provide clinically actionable insights for disease diagnosis, prognosis, and treatment. Computational methods aid the discovery by relying on protein-protein interaction (PPI) networks. They start with a few known disease-associated proteins and aim to find the rest of the pathway by exploring the PPI network around the known disease proteins. However, the success of such methods has been limited, and failure cases have not been well understood. Here we study the PPI network structure of 519 disease pathways. We find that 90% of pathways do not correspond to single well-connected components in the PPI network. Instead, proteins associated with a single disease tend to form many separate connected components/regions in the network. We then evaluate state-of-the-art disease pathway discovery methods and show that their performance is especially poor on diseases with disconnected pathways. Thus, we conclude that network connectivity structure alone may not be sufficient for disease pathway discovery. However, we show that higher-order network structures, such as small subgraphs of the pathway, provide a promising direction for the development of new methods.


Subject(s)
Disease/etiology , Protein Interaction Maps , Algorithms , Computational Biology/methods , Humans , Protein Interaction Mapping/methods , Protein Interaction Mapping/statistics & numerical data , Proteome , Proteomics/methods , Proteomics/statistics & numerical data , Signal Transduction
20.
PLoS Comput Biol ; 13(12): e1005905, 2017 12.
Article in English | MEDLINE | ID: mdl-29281622

ABSTRACT

Peptide-protein interactions contribute a significant fraction of the protein-protein interactome. Accurate modeling of these interactions is challenging due to the vast conformational space associated with interactions of highly flexible peptides with large receptor surfaces. To address this challenge we developed a fragment based high-resolution peptide-protein docking protocol. By streamlining the Rosetta fragment picker for accurate peptide fragment ensemble generation, the PIPER docking algorithm for exhaustive fragment-receptor rigid-body docking and Rosetta FlexPepDock for flexible full-atom refinement of PIPER docked models, we successfully addressed the challenge of accurate and efficient global peptide-protein docking at high-resolution with remarkable accuracy, as validated on a small but representative set of peptide-protein complex structures well resolved by X-ray crystallography. Our approach opens up the way to high-resolution modeling of many more peptide-protein interactions and to the detailed study of peptide-protein association in general. PIPER-FlexPepDock is freely available to the academic community as a server at http://piperfpd.furmanlab.cs.huji.ac.il.


Subject(s)
Algorithms , Protein Interaction Mapping/statistics & numerical data , Computational Biology , Computer Simulation , Crystallography, X-Ray , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Protein Conformation , Software
SELECTION OF CITATIONS
SEARCH DETAIL