Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
mSystems ; 7(2): e0146621, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35319251

ABSTRACT

Suppression of the host innate immune response is a critical aspect of viral replication. Upon infection, viruses may introduce one or more proteins that inhibit key immune pathways, such as the type I interferon pathway. However, the ability to predict and evaluate viral protein bioactivity on targeted pathways remains challenging and is typically done on a single-virus or -gene basis. Here, we present a medium-throughput high-content cell-based assay to reveal the immunosuppressive effects of viral proteins. To test the predictive power of our approach, we developed a library of 800 genes encoding known, predicted, and uncharacterized human virus genes. We found that previously known immune suppressors from numerous viral families such as Picornaviridae and Flaviviridae recorded positive responses. These include a number of viral proteases for which we further confirmed that innate immune suppression depends on protease activity. A class of predicted inhibitors encoded by Rhabdoviridae viruses was demonstrated to block nuclear transport, and several previously uncharacterized proteins from uncultivated viruses were shown to inhibit nuclear transport of the transcription factors NF-κB and interferon regulatory factor 3 (IRF3). We propose that this pathway-based assay, together with early sequencing, gene synthesis, and viral infection studies, could partly serve as the basis for rapid in vitro characterization of novel viral proteins. IMPORTANCE Infectious diseases caused by viral pathogens exacerbate health care and economic burdens. Numerous viral biomolecules suppress the human innate immune system, enabling viruses to evade an immune response from the host. Despite our current understanding of viral replications and immune evasion, new viral proteins, including those encoded by uncultivated viruses or emerging viruses, are being unearthed at a rapid pace from large-scale sequencing and surveillance projects. The use of medium- and high-throughput functional assays to characterize immunosuppressive functions of viral proteins can advance our understanding of viral replication and possibly treatment of infections. In this study, we assembled a large viral-gene library from diverse viral families and developed a high-content assay to test for inhibition of innate immunity pathways. Our work expands the tools that can rapidly link sequence and protein function, representing a practical step toward early-stage evaluation of emerging and understudied viruses.


Subject(s)
Immunity, Innate , Viruses , Humans , NF-kappa B , Immune Evasion , Viruses/genetics , Viral Proteins/genetics , Genes, Viral
2.
ACS Synth Biol ; 11(3): 1292-1302, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35176859

ABSTRACT

Many organisms can survive extreme conditions and successfully recover to normal life. This extremotolerant behavior has been attributed in part to repetitive, amphipathic, and intrinsically disordered proteins that are upregulated in the protected state. Here, we assemble a library of approximately 300 naturally occurring and designed extremotolerance-associated proteins to assess their ability to protect human cells from chemically induced apoptosis. We show that several proteins from tardigrades, nematodes, and the Chinese giant salamander are apoptosis-protective. Notably, we identify a region of the human ApoE protein with similarity to extremotolerance-associated proteins that also protects against apoptosis. This region mirrors the phase separation behavior seen with such proteins, like the tardigrade protein CAHS2. Moreover, we identify a synthetic protein, DHR81, that shares this combination of elevated phase separation propensity and apoptosis protection. Finally, we demonstrate that driving protective proteins into the condensate state increases apoptosis protection, and highlights the ability of DHR81 condensates to sequester caspase-7. Taken together, this work draws a link between extremotolerance-associated proteins, condensate formation, and designing human cellular protection.


Subject(s)
Intrinsically Disordered Proteins , Tardigrada , Animals , Apoptosis , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Tardigrada/metabolism
3.
EMBO J ; 39(23): e104523, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33073387

ABSTRACT

Oxidative stress alters cell viability, from microorganism irradiation sensitivity to human aging and neurodegeneration. Deleterious effects of protein carbonylation by reactive oxygen species (ROS) make understanding molecular properties determining ROS susceptibility essential. The radiation-resistant bacterium Deinococcus radiodurans accumulates less carbonylation than sensitive organisms, making it a key model for deciphering properties governing oxidative stress resistance. We integrated shotgun redox proteomics, structural systems biology, and machine learning to resolve properties determining protein damage by γ-irradiation in Escherichia coli and D. radiodurans at multiple scales. Local accessibility, charge, and lysine enrichment accurately predict ROS susceptibility. Lysine, methionine, and cysteine usage also contribute to ROS resistance of the D. radiodurans proteome. Our model predicts proteome maintenance machinery, and proteins protecting against ROS are more resistant in D. radiodurans. Our findings substantiate that protein-intrinsic protection impacts oxidative stress resistance, identifying causal molecular properties.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Oxidative Stress/physiology , Proteome/metabolism , Aging/metabolism , Computational Biology , Deinococcus/metabolism , Escherichia coli , Humans , Machine Learning , Neurodegenerative Diseases/metabolism , Oxidation-Reduction , Protein Conformation , Protein Processing, Post-Translational , Proteomics/methods , Reactive Oxygen Species/metabolism , Sequence Analysis, Protein
4.
Proc Natl Acad Sci U S A ; 115(43): 11096-11101, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30301795

ABSTRACT

Understanding the complex interactions of protein posttranslational modifications (PTMs) represents a major challenge in metabolic engineering, synthetic biology, and the biomedical sciences. Here, we present a workflow that integrates multiplex automated genome editing (MAGE), genome-scale metabolic modeling, and atomistic molecular dynamics to study the effects of PTMs on metabolic enzymes and microbial fitness. This workflow incorporates complementary approaches across scientific disciplines; provides molecular insight into how PTMs influence cellular fitness during nutrient shifts; and demonstrates how mechanistic details of PTMs can be explored at different biological scales. As a proof of concept, we present a global analysis of PTMs on enzymes in the metabolic network of Escherichia coli Based on our workflow results, we conduct a more detailed, mechanistic analysis of the PTMs in three proteins: enolase, serine hydroxymethyltransferase, and transaldolase. Application of this workflow identified the roles of specific PTMs in observed experimental phenomena and demonstrated how individual PTMs regulate enzymes, pathways, and, ultimately, cell phenotypes.


Subject(s)
Prokaryotic Cells/metabolism , Protein Processing, Post-Translational/genetics , Escherichia coli/metabolism , Gene Editing/methods , Metabolic Engineering/methods , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Workflow
5.
Mol Biosyst ; 12(8): 2394-407, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27357594

ABSTRACT

Metabolic networks, which are mathematical representations of organismal metabolism, are reconstructed to provide computational platforms to guide metabolic engineering experiments and explore fundamental questions on metabolism. Systems level analyses, such as interrogation of phylogenetic relationships within the network, can provide further guidance on the modification of metabolic circuitries. Chlamydomonas reinhardtii, a biofuel relevant green alga that has retained key genes with plant, animal, and protist affinities, serves as an ideal model organism to investigate the interplay between gene function and phylogenetic affinities at multiple organizational levels. Here, using detailed topological and functional analyses, coupled with transcriptomics studies on a metabolic network that we have reconstructed for C. reinhardtii, we show that network connectivity has a significant concordance with the co-conservation of genes; however, a distinction between topological and functional relationships is observable within the network. Dynamic and static modes of co-conservation were defined and observed in a subset of gene-pairs across the network topologically. In contrast, genes with predicted synthetic interactions, or genes involved in coupled reactions, show significant enrichment for both shorter and longer phylogenetic distances. Based on our results, we propose that the metabolic network of C. reinhardtii is assembled with an architecture to minimize phylogenetic profile distances topologically, while it includes an expansion of such distances for functionally interacting genes. This arrangement may increase the robustness of C. reinhardtii's network in dealing with varied environmental challenges that the species may face. The defined evolutionary constraints within the network, which identify important pairings of genes in metabolism, may offer guidance on synthetic biology approaches to optimize the production of desirable metabolites.


Subject(s)
Biological Evolution , Chlamydomonas reinhardtii/metabolism , Metabolic Networks and Pathways , Synthetic Biology , Chlamydomonas reinhardtii/genetics , Computational Biology/methods , Evolution, Molecular , Gene Ontology , Gene Regulatory Networks , Genomics/methods , Open Reading Frames/genetics , Synthetic Biology/methods
6.
BMC Syst Biol ; 10: 26, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26969117

ABSTRACT

BACKGROUND: The success of genome-scale models (GEMs) can be attributed to the high-quality, bottom-up reconstructions of metabolic, protein synthesis, and transcriptional regulatory networks on an organism-specific basis. Such reconstructions are biochemically, genetically, and genomically structured knowledge bases that can be converted into a mathematical format to enable a myriad of computational biological studies. In recent years, genome-scale reconstructions have been extended to include protein structural information, which has opened up new vistas in systems biology research and empowered applications in structural systems biology and systems pharmacology. RESULTS: Here, we present the generation, application, and dissemination of genome-scale models with protein structures (GEM-PRO) for Escherichia coli and Thermotoga maritima. We show the utility of integrating molecular scale analyses with systems biology approaches by discussing several comparative analyses on the temperature dependence of growth, the distribution of protein fold families, substrate specificity, and characteristic features of whole cell proteomes. Finally, to aid in the grand challenge of big data to knowledge, we provide several explicit tutorials of how protein-related information can be linked to genome-scale models in a public GitHub repository ( https://github.com/SBRG/GEMPro/tree/master/GEMPro_recon/). CONCLUSIONS: Translating genome-scale, protein-related information to structured data in the format of a GEM provides a direct mapping of gene to gene-product to protein structure to biochemical reaction to network states to phenotypic function. Integration of molecular-level details of individual proteins, such as their physical, chemical, and structural properties, further expands the description of biochemical network-level properties, and can ultimately influence how to model and predict whole cell phenotypes as well as perform comparative systems biology approaches to study differences between organisms. GEM-PRO offers insight into the physical embodiment of an organism's genotype, and its use in this comparative framework enables exploration of adaptive strategies for these organisms, opening the door to many new lines of research. With these provided tools, tutorials, and background, the reader will be in a position to run GEM-PRO for their own purposes.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Proteomics , Systems Biology/methods , Thermotoga maritima/genetics , Thermotoga maritima/metabolism , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Biological , Models, Molecular , Protein Conformation , Sequence Homology, Amino Acid , Temperature , Thermotoga maritima/growth & development
7.
Sci Rep ; 6: 20086, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26833023

ABSTRACT

Streptomyces thermoautotrophicus UBT1 has been described as a moderately thermophilic chemolithoautotroph with a novel nitrogenase enzyme that is oxygen-insensitive. We have cultured the UBT1 strain, and have isolated two new strains (H1 and P1-2) of very similar phenotypic and genetic characters. These strains show minimal growth on ammonium-free media, and fail to incorporate isotopically labeled N2 gas into biomass in multiple independent assays. The sdn genes previously published as the putative nitrogenase of S. thermoautotrophicus have little similarity to anything found in draft genome sequences, published here, for strains H1 and UBT1, but share >99% nucleotide identity with genes from Hydrogenibacillus schlegelii, a draft genome for which is also presented here. H. schlegelii similarly lacks nitrogenase genes and is a non-diazotroph. We propose reclassification of the species containing strains UBT1, H1, and P1-2 as a non-Streptomycete, non-diazotrophic, facultative chemolithoautotroph and conclude that the existence of the previously proposed oxygen-tolerant nitrogenase is extremely unlikely.


Subject(s)
Genes, Bacterial , Nitrogen Fixation , Streptomyces/genetics , Streptomyces/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Isotope Labeling , Nitrogen/metabolism , Nitrogenase/genetics , Nitrogenase/metabolism , Sequence Homology, Nucleic Acid
9.
Sci Adv ; 1(5)2015.
Article in English | MEDLINE | ID: mdl-26229984

ABSTRACT

Circadian oscillators are posttranslationally regulated and affect gene expression in autotrophic cyanobacteria. Oscillations are controlled by phosphorylation of the KaiC protein, which is modulated by the KaiA and KaiB proteins. However, it remains unclear how time information is transmitted to transcriptional output. We show reconstruction of the KaiABC oscillator in the noncircadian bacterium Escherichia coli. This orthogonal system shows circadian oscillations in KaiC phosphorylation and in a synthetic transcriptional reporter. Coexpression of KaiABC with additional native cyanobacterial components demonstrates a minimally sufficient set of proteins for transcriptional output from a native cyanobacterial promoter in E. coli. Together, these results demonstrate that a circadian oscillator is transplantable to a heterologous organism for reductive study as well as wide-ranging applications.

10.
Mol Syst Biol ; 9: 693, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24084808

ABSTRACT

Growth is a fundamental process of life. Growth requirements are well-characterized experimentally for many microbes; however, we lack a unified model for cellular growth. Such a model must be predictive of events at the molecular scale and capable of explaining the high-level behavior of the cell as a whole. Here, we construct an ME-Model for Escherichia coli--a genome-scale model that seamlessly integrates metabolic and gene product expression pathways. The model computes ~80% of the functional proteome (by mass), which is used by the cell to support growth under a given condition. Metabolism and gene expression are interdependent processes that affect and constrain each other. We formalize these constraints and apply the principle of growth optimization to enable the accurate prediction of multi-scale phenotypes, ranging from coarse-grained (growth rate, nutrient uptake, by-product secretion) to fine-grained (metabolic fluxes, gene expression levels). Our results unify many existing principles developed to describe bacterial growth.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Genome, Bacterial , Metabolic Networks and Pathways/genetics , Models, Biological , Escherichia coli/growth & development , Escherichia coli/metabolism , Genetic Association Studies , Genotype , Phenotype
11.
BMC Syst Biol ; 7: 102, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24112686

ABSTRACT

BACKGROUND: The growing discipline of structural systems pharmacology is applied prospectively in this study to predict pharmacological outcomes of antibacterial compounds in Escherichia coli K12. This work builds upon previously established methods for structural prediction of ligand binding pockets on protein molecules and utilizes and expands upon the previously developed genome scale model of metabolism integrated with protein structures (GEM-PRO) for E. coli, structurally accounting for protein complexes. Carefully selected case studies are demonstrated to display the potential for this structural systems pharmacology framework in discovery and development of antibacterial compounds. RESULTS: The prediction framework for antibacterial activity of compounds was validated for a control set of well-studied compounds, recapitulating experimentally-determined protein binding interactions and deleterious growth phenotypes resulting from these interactions. The antibacterial activity of fosfomycin, sulfathiazole, and trimethoprim were accurately predicted, and as a negative control glucose was found to have no predicted antibacterial activity. Previously uncharacterized mechanisms of action were predicted for compounds with known antibacterial properties, including (1-hydroxyheptane-1,1-diyl)bis(phosphonic acid) and cholesteryl oleate. Five candidate inhibitors were predicted for a desirable target protein without any known inhibitors, tryptophan synthase ß subunit (TrpB). In addition to the predictions presented, this effort also included significant expansion of the previously developed GEM-PRO to account for physiological assemblies of protein complex structures with activities included in the E. coli K12 metabolic network. CONCLUSIONS: The structural systems pharmacology framework presented in this study was shown to be effective in the prediction of molecular mechanisms of antibacterial compounds. The study provides a promising proof of principle for such an approach to antibacterial development and raises specific molecular and systemic hypotheses about antibacterials that are amenable to experimental testing. This framework, and perhaps also the specific predictions of antibacterials, is extensible to developing antibacterial treatments for pathogenic E. coli and other bacterial pathogens.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Systems Biology/methods , Anti-Bacterial Agents/metabolism , Binding Sites , Escherichia coli K12/drug effects , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Genomics , Models, Molecular , Phenotype , Protein Conformation
12.
Science ; 340(6137): 1220-3, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23744946

ABSTRACT

Genome-scale network reconstruction has enabled predictive modeling of metabolism for many systems. Traditionally, protein structural information has not been represented in such reconstructions. Expansion of a genome-scale model of Escherichia coli metabolism by including experimental and predicted protein structures enabled the analysis of protein thermostability in a network context. This analysis allowed the prediction of protein activities that limit network function at superoptimal temperatures and mechanistic interpretations of mutations found in strains adapted to heat. Predicted growth-limiting factors for thermotolerance were validated through nutrient supplementation experiments and defined metabolic sensitivities to heat stress, providing evidence that metabolic enzyme thermostability is rate-limiting at superoptimal temperatures. Inclusion of structural information expanded the content and predictive capability of genome-scale metabolic networks that enable structural systems biology of metabolism.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Hot Temperature , Metabolic Networks and Pathways , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Models, Biological , Protein Conformation , Systems Biology , Transcriptional Activation
13.
Science ; 337(6098): 1101-4, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22936779

ABSTRACT

Enzymes are thought to have evolved highly specific catalytic activities from promiscuous ancestral proteins. By analyzing a genome-scale model of Escherichia coli metabolism, we found that 37% of its enzymes act on a variety of substrates and catalyze 65% of the known metabolic reactions. However, it is not apparent why these generalist enzymes remain. Here, we show that there are marked differences between generalist enzymes and specialist enzymes, known to catalyze a single chemical reaction on one particular substrate in vivo. Specialist enzymes (i) are frequently essential, (ii) maintain higher metabolic flux, and (iii) require more regulation of enzyme activity to control metabolic flux in dynamic environments than do generalist enzymes. Furthermore, these properties are conserved in Archaea and Eukarya. Thus, the metabolic network context and environmental conditions influence enzyme evolution toward high specificity.


Subject(s)
Enzymes/genetics , Enzymes/metabolism , Escherichia coli/enzymology , Evolution, Molecular , Metabolic Networks and Pathways , Selection, Genetic , Catalysis , Computational Biology , Escherichia coli/genetics , Substrate Specificity
14.
Mol Syst Biol ; 7: 518, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21811229

ABSTRACT

Metabolic network reconstruction encompasses existing knowledge about an organism's metabolism and genome annotation, providing a platform for omics data analysis and phenotype prediction. The model alga Chlamydomonas reinhardtii is employed to study diverse biological processes from photosynthesis to phototaxis. Recent heightened interest in this species results from an international movement to develop algal biofuels. Integrating biological and optical data, we reconstructed a genome-scale metabolic network for this alga and devised a novel light-modeling approach that enables quantitative growth prediction for a given light source, resolving wavelength and photon flux. We experimentally verified transcripts accounted for in the network and physiologically validated model function through simulation and generation of new experimental growth data, providing high confidence in network contents and predictive applications. The network offers insight into algal metabolism and potential for genetic engineering and efficient light source design, a pioneering resource for studying light-driven metabolism and quantitative systems biology.


Subject(s)
Chlamydomonas reinhardtii/genetics , Chlamydomonas reinhardtii/metabolism , Metabolic Networks and Pathways/genetics , Chlamydomonas reinhardtii/growth & development , Computer Simulation , Databases, Genetic , Genetic Engineering , Lipid Metabolism , Models, Biological , Phenotype , Photobioreactors , Photosynthesis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Systems Biology/methods
15.
PLoS Comput Biol ; 6(9): e1000938, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20957118

ABSTRACT

Recent advances in structural bioinformatics have enabled the prediction of protein-drug off-targets based on their ligand binding sites. Concurrent developments in systems biology allow for prediction of the functional effects of system perturbations using large-scale network models. Integration of these two capabilities provides a framework for evaluating metabolic drug response phenotypes in silico. This combined approach was applied to investigate the hypertensive side effect of the cholesteryl ester transfer protein inhibitor torcetrapib in the context of human renal function. A metabolic kidney model was generated in which to simulate drug treatment. Causal drug off-targets were predicted that have previously been observed to impact renal function in gene-deficient patients and may play a role in the adverse side effects observed in clinical trials. Genetic risk factors for drug treatment were also predicted that correspond to both characterized and unknown renal metabolic disorders as well as cryptic genetic deficiencies that are not expected to exhibit a renal disorder phenotype except under drug treatment. This study represents a novel integration of structural and systems biology and a first step towards computational systems medicine. The methodology introduced herein has important implications for drug development and personalized medicine.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Kidney/drug effects , Kidney/metabolism , Metabolic Networks and Pathways/drug effects , Models, Biological , Algorithms , Gene Expression Profiling , Humans , Kidney Diseases/metabolism , Kidney Function Tests , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/physiology , Metabolomics , Protein Binding , Proteome/drug effects , Proteome/genetics , Proteome/metabolism , Quinolines/pharmacology , ROC Curve
16.
Int J Bioinform Res Appl ; 6(2): 101-19, 2010.
Article in English | MEDLINE | ID: mdl-20223734

ABSTRACT

An approach for module identification, Modules of Networks (MoNet), introduced an intuitive module definition and clear detection method using edges ranked by the Girvan-Newman algorithm. Modules from a yeast network showed significant association with biological processes, indicating the method's utility; however, systematic bias leads to varied results across trials. MoNet modules also exclude some network regions. To address these shortcomings, we developed a deterministic version of the Girvan-Newman algorithm and a new agglomerative algorithm, Deterministic Modularization of Networks (dMoNet). dMoNet simultaneously processes structurally equivalent edges while preserving intuitive foundations of the MoNet algorithm and generates modules with full network coverage.


Subject(s)
Algorithms , Proteome/analysis , Proteomics/methods , Protein Interaction Mapping/methods
17.
Nat Methods ; 6(8): 589-92, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597503

ABSTRACT

With sequencing of thousands of organisms completed or in progress, there is a growing need to integrate gene prediction with metabolic network analysis. Using Chlamydomonas reinhardtii as a model, we describe a systems-level methodology bridging metabolic network reconstruction with experimental verification of enzyme encoding open reading frames. Our quantitative and predictive metabolic model and its associated cloned open reading frames provide useful resources for metabolic engineering.


Subject(s)
Chlamydomonas reinhardtii/metabolism , Computational Biology/methods , Genome, Protozoan , Models, Genetic , Protozoan Proteins/metabolism , Transcription, Genetic , Animals , Chlamydomonas reinhardtii/enzymology , Chlamydomonas reinhardtii/genetics , Computer Simulation , Enzymes/genetics , Enzymes/metabolism , Genetic Engineering , Protozoan Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...