Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 1267, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741116

ABSTRACT

Proliferation of multidrug-resistant (MDR) bacteria poses a threat to human health, requiring new strategies. Here we propose using fitness neutral gene expression perturbations to potentiate antibiotics. We systematically explored 270 gene knockout-antibiotic combinations in Escherichia coli, identifying 90 synergistic interactions. Identified gene targets were subsequently tested for antibiotic synergy on the transcriptomic level via multiplexed CRISPR-dCas9 and showed successful sensitization of E. coli without a separate fitness cost. These fitness neutral gene perturbations worked as co-therapies in reducing a Salmonella enterica intracellular infection in HeLa. Finally, these results informed the design of four antisense peptide nucleic acid (PNA) co-therapies, csgD, fnr, recA and acrA, against four MDR, clinically isolated bacteria. PNA combined with sub-minimal inhibitory concentrations of trimethoprim against two isolates of Klebsiella pneumoniae and E. coli showed three cases of re-sensitization with minimal fitness impacts. Our results highlight a promising approach for extending the utility of current antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Gene Expression/drug effects , Klebsiella pneumoniae/genetics , Salmonella enterica/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Salmonella enterica/drug effects
2.
Commun Biol ; 4(1): 331, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712689

ABSTRACT

Multidrug-resistant (MDR) bacteria pose a grave concern to global health, which is perpetuated by a lack of new treatments and countermeasure platforms to combat outbreaks or antibiotic resistance. To address this, we have developed a Facile Accelerated Specific Therapeutic (FAST) platform that can develop effective peptide nucleic acid (PNA) therapies against MDR bacteria within a week. Our FAST platform uses a bioinformatics toolbox to design sequence-specific PNAs targeting non-traditional pathways/genes of bacteria, then performs in-situ synthesis, validation, and efficacy testing of selected PNAs. As a proof of concept, these PNAs were tested against five MDR clinical isolates: carbapenem-resistant Escherichia coli, extended-spectrum beta-lactamase Klebsiella pneumoniae, New Delhi Metallo-beta-lactamase-1 carrying Klebsiella pneumoniae, and MDR Salmonella enterica. PNAs showed significant growth inhibition for 82% of treatments, with nearly 18% of treatments leading to greater than 97% decrease. Further, these PNAs are capable of potentiating antibiotic activity in the clinical isolates despite presence of cognate resistance genes. Finally, the FAST platform offers a novel delivery approach to overcome limited transport of PNAs into mammalian cells by repurposing the bacterial Type III secretion system in conjunction with a kill switch that is effective at eliminating 99.6% of an intracellular Salmonella infection in human epithelial cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Computational Biology , Drug Design , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae/drug effects , Oligonucleotides, Antisense/pharmacology , Peptide Nucleic Acids/pharmacology , 3T3 Cells , Animals , Drug Resistance, Multiple, Bacterial/genetics , Enterobacteriaceae/genetics , Enterobacteriaceae/growth & development , Enterobacteriaceae Infections/microbiology , HeLa Cells , Humans , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Proof of Concept Study , RAW 264.7 Cells
3.
Proc Natl Acad Sci U S A ; 117(48): 30699-30709, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33199638

ABSTRACT

In recent years, the prevalence of carbapenem-resistant Enterobacteriaceae (CRE) has risen substantially, and the study of CRE resistance mechanisms has become increasingly important for antibiotic development. Although much research has focused on genomic resistance factors, relatively few studies have examined CRE pathogens through changes in gene expression. In this study, we examined the gene expression profile of a CRE Escherichia coli clinical isolate that is sensitive to meropenem but resistant to ertapenem to explore transcriptomic contributions to resistance and to identify gene knockdown targets for carbapenem potentiation. We sequenced total and short RNA to analyze the gene expression response to ertapenem or meropenem treatment and found significant expression changes in genes related to motility, maltodextrin metabolism, the formate hydrogenlyase complex, and the general stress response. To validate these findings, we used our laboratory's Facile Accelerated Specific Therapeutic (FAST) platform to create antisense peptide nucleic acids (PNAs), gene-specific molecules designed to inhibit protein translation. PNAs were designed to inhibit the pathways identified in our transcriptomic analysis, and each PNA was then tested in combination with each carbapenem to assess its effect on the antibiotics' minimum inhibitory concentrations. We observed significant PNA-antibiotic interaction with five different PNAs across six combinations. Inhibition of the genes hycA, dsrB, and bolA potentiated carbapenem efficacy in CRE E. coli, whereas inhibition of the genes flhC and ygaC conferred added resistance. Our results identify resistance factors and demonstrate that transcriptomic analysis is a potent tool for designing antibiotic PNA.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Carbapenem-Resistant Enterobacteriaceae/genetics , Carbapenems/pharmacology , Gene Expression Profiling , Oligonucleotides, Antisense , Transcriptome , Anti-Bacterial Agents/chemistry , Carbapenems/chemistry , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae Infections/microbiology , Gene Expression Profiling/methods , Genome, Bacterial , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Meropenem/pharmacology , Microbial Sensitivity Tests
4.
Methods Mol Biol ; 1945: 391-419, 2019.
Article in English | MEDLINE | ID: mdl-30945257

ABSTRACT

BioNetFit is a software tool designed for solving parameter identification problems that arise in the development of rule-based models. It solves these problems through curve fitting (i.e., nonlinear regression). BioNetFit is compatible with deterministic and stochastic simulators that accept BioNetGen language (BNGL)-formatted files as inputs, such as those available within the BioNetGen framework. BioNetFit can be used on a laptop or stand-alone multicore workstation as well as on many Linux clusters, such as those that use the Slurm Workload Manager to schedule jobs. BioNetFit implements a metaheuristic population-based global optimization procedure, an evolutionary algorithm (EA), to minimize a user-defined objective function, such as a residual sum of squares (RSS) function. BioNetFit also implements a bootstrapping procedure for determining confidence intervals for parameter estimates. Here, we provide step-by-step instructions for using BioNetFit to estimate the values of parameters of a BNGL-encoded model and to define bootstrap confidence intervals. The process entails the use of several plain-text files, which are processed by BioNetFit and BioNetGen. In general, these files include (1) one or more EXP files, which each contains (experimental) data to be used in parameter identification/bootstrapping; (2) a BNGL file containing a model section, which defines a (rule-based) model, and an actions section, which defines simulation protocols that generate GDAT and/or SCAN files with model predictions corresponding to the data in the EXP file(s); and (3) a CONF file that configures the fitting/bootstrapping job and that defines algorithmic parameter settings.


Subject(s)
Computational Biology/methods , Models, Biological , Software , Systems Biology/methods , Algorithms , Computer Simulation
5.
PLoS Comput Biol ; 15(1): e1006706, 2019 01.
Article in English | MEDLINE | ID: mdl-30653502

ABSTRACT

Receptor tyrosine kinases (RTKs) typically contain multiple autophosphorylation sites in their cytoplasmic domains. Once activated, these autophosphorylation sites can recruit downstream signaling proteins containing Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains, which recognize phosphotyrosine-containing short linear motifs (SLiMs). These domains and SLiMs have polyspecific or promiscuous binding activities. Thus, multiple signaling proteins may compete for binding to a common SLiM and vice versa. To investigate the effects of competition on RTK signaling, we used a rule-based modeling approach to develop and analyze models for ligand-induced recruitment of SH2/PTB domain-containing proteins to autophosphorylation sites in the insulin-like growth factor 1 (IGF1) receptor (IGF1R). Models were parameterized using published datasets reporting protein copy numbers and site-specific binding affinities. Simulations were facilitated by a novel application of model restructuration, to reduce redundancy in rule-derived equations. We compare predictions obtained via numerical simulation of the model to those obtained through simple prediction methods, such as through an analytical approximation, or ranking by copy number and/or KD value, and find that the simple methods are unable to recapitulate the predictions of numerical simulations. We created 45 cell line-specific models that demonstrate how early events in IGF1R signaling depend on the protein abundance profile of a cell. Simulations, facilitated by model restructuration, identified pairs of IGF1R binding partners that are recruited in anti-correlated and correlated fashions, despite no inclusion of cooperativity in our models. This work shows that the outcome of competition depends on the physicochemical parameters that characterize pairwise interactions, as well as network properties, including network connectivity and the relative abundances of competitors.


Subject(s)
Models, Biological , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Binding Sites , Cell Line , Cluster Analysis , Computational Biology , Humans , Mice , Phosphorylation , Protein Binding , Proteins/chemistry , Proteins/metabolism , src Homology Domains
6.
Semin Cancer Biol ; 54: 162-173, 2019 02.
Article in English | MEDLINE | ID: mdl-29518522

ABSTRACT

RAS is the most frequently mutated gene across human cancers, but developing inhibitors of mutant RAS has proven to be challenging. Given the difficulties of targeting RAS directly, drugs that impact the other components of pathways where mutant RAS operates may potentially be effective. However, the system-level features, including different localizations of RAS isoforms, competition between downstream effectors, and interlocking feedback and feed-forward loops, must be understood to fully grasp the opportunities and limitations of inhibiting specific targets. Mathematical modeling can help us discern the system-level impacts of these features in normal and cancer cells. New technologies enable the acquisition of experimental data that will facilitate development of realistic models of oncogenic RAS behavior. In light of the wealth of empirical data accumulated over decades of study and the advancement of experimental methods for gathering new data, modelers now have the opportunity to advance progress toward realization of targeted treatment for mutant RAS-driven cancers.


Subject(s)
Gene Expression Regulation , Models, Biological , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism , Animals , Carrier Proteins , Drug Discovery , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Protein Transport , Systems Biology/methods , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry
7.
Bull Math Biol ; 81(8): 2822-2848, 2019 08.
Article in English | MEDLINE | ID: mdl-29594824

ABSTRACT

Gillespie's direct method for stochastic simulation of chemical kinetics is a staple of computational systems biology research. However, the algorithm requires explicit enumeration of all reactions and all chemical species that may arise in the system. In many cases, this is not feasible due to the combinatorial explosion of reactions and species in biological networks. Rule-based modeling frameworks provide a way to exactly represent networks containing such combinatorial complexity, and generalizations of Gillespie's direct method have been developed as simulation engines for rule-based modeling languages. Here, we provide both a high-level description of the algorithms underlying the simulation engines, termed network-free simulation algorithms, and how they have been applied in systems biology research. We also define a generic rule-based modeling framework and describe a number of technical details required for adapting Gillespie's direct method for network-free simulation. Finally, we briefly discuss potential avenues for advancing network-free simulation and the role they continue to play in modeling dynamical systems in biology.


Subject(s)
Algorithms , Computer Simulation , Systems Biology/methods , Biochemical Phenomena , Kinetics , Mathematical Concepts , Metabolic Networks and Pathways , Models, Biological , Monte Carlo Method , Stochastic Processes , Terminology as Topic
8.
Cell Syst ; 7(2): 161-179.e14, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30007540

ABSTRACT

Clinically used RAF inhibitors are ineffective in RAS mutant tumors because they enhance homo- and heterodimerization of RAF kinases, leading to paradoxical activation of ERK signaling. Overcoming enhanced RAF dimerization and the resulting resistance is a challenge for drug design. Combining multiple inhibitors could be more effective, but it is unclear how the best combinations can be chosen. We built a next-generation mechanistic dynamic model to analyze combinations of structurally different RAF inhibitors, which can efficiently suppress MEK/ERK signaling. This rule-based model of the RAS/ERK pathway integrates thermodynamics and kinetics of drug-protein interactions, structural elements, posttranslational modifications, and cell mutational status as model rules to predict RAF inhibitor combinations for inhibiting ERK activity in oncogenic RAS and/or BRAFV600E backgrounds. Predicted synergistic inhibition of ERK signaling was corroborated by experiments in mutant NRAS, HRAS, and BRAFV600E cells, and inhibition of oncogenic RAS signaling was associated with reduced cell proliferation and colony formation.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , ras Proteins/metabolism , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Molecular Docking Simulation , Mutation/drug effects , Neoplasms/genetics , Neoplasms/metabolism , Protein Multimerization/drug effects , Thermodynamics , raf Kinases/chemistry , raf Kinases/metabolism , ras Proteins/genetics
9.
Front Microbiol ; 9: 310, 2018.
Article in English | MEDLINE | ID: mdl-29615983

ABSTRACT

Bacteria grown in space experiments under microgravity conditions have been found to undergo unique physiological responses, ranging from modified cell morphology and growth dynamics to a putative increased tolerance to antibiotics. A common theory for this behavior is the loss of gravity-driven convection processes in the orbital environment, resulting in both reduction of extracellular nutrient availability and the accumulation of bacterial byproducts near the cell. To further characterize the responses, this study investigated the transcriptomic response of Escherichia coli to both microgravity and antibiotic concentration. E. coli was grown aboard International Space Station in the presence of increasing concentrations of the antibiotic gentamicin with identical ground controls conducted on Earth. Here we show that within 49 h of being cultured, E. coli adapted to grow at higher antibiotic concentrations in space compared to Earth, and demonstrated consistent changes in expression of 63 genes in response to an increase in drug concentration in both environments, including specific responses related to oxidative stress and starvation response. Additionally, we find 50 stress-response genes upregulated in response to the microgravity when compared directly to the equivalent concentration in the ground control. We conclude that the increased antibiotic tolerance in microgravity may be attributed not only to diminished transport processes, but also to a resultant antibiotic cross-resistance response conferred by an overlapping effect of stress response genes. Our data suggest that direct stresses of nutrient starvation and acid-shock conveyed by the microgravity environment can incidentally upregulate stress response pathways related to antibiotic stress and in doing so contribute to the increased antibiotic stress tolerance observed for bacteria in space experiments. These results provide insights into the ability of bacteria to adapt under extreme stress conditions and potential strategies to prevent antimicrobial-resistance in space and on Earth.

10.
ACS Synth Biol ; 6(12): 2302-2315, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29017328

ABSTRACT

Tolerance and resistance are complex biological phenotypes that are desirable bioengineering goals for those seeking to design industrial strains or prevent the spread of antibiotic resistance. Over decades of research, a wealth of information has been generated to attempt to decode a molecular basis for tolerance, but to fully achieve the goal of engineering tolerance, researchers must be able to easily learn from a variety of data sources. To this end, we here describe a resource designed to enable scrutiny of diverse tolerance phenotypes. We have curated hundreds of gene expression studies exploring the response of Escherichia coli to chemical and environmental perturbations, from antibiotics to biofuels and solvents and more. Overall, our efforts give rise to a database encompassing more than 56 000 gene expression changes across 89 different stress conditions. This resource is designed for compatibility with the Resistome database, which includes more than 5000 strains with mutations conferring resistance or sensitivity but no transcriptomic data. Thus, the work here results in the first combined resource specialized to tolerance and resistance in E. coli that supports investigations across genomic, transcriptomic, and phenotypic levels. We leverage the database to identify promising bioengineering targets by searching globally across multiple stress conditions as well as by narrowing the focus to fewer conditions of interest, such as biofuel stress and antibiotic stress. We discuss some of the most frequently differentially expressed or coexpressed genes, and predict which transcription factors and sigma factors most likely contribute to gene expression profiles in a wide array of conditions. We also compare profiles from sensitive and resistant strains, gaining knowledge of how responses differ per overrepresented gene ontology terms. Finally, we search for genes that are frequently differentially expressed but not mutated, with the expectation that these may present interesting targets for future engineering efforts. The curated data presented here is publicly available, and should be advantageous to those studying a variety of bacterial tolerance phenotypes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Databases, Nucleic Acid , Drug Resistance, Bacterial , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Anti-Bacterial Agents/chemistry
11.
Biotechnol Bioeng ; 114(11): 2685-2689, 2017 11.
Article in English | MEDLINE | ID: mdl-28710857

ABSTRACT

The economical production of chemicals and fuels by microbial processes remains an intense area of interest in biotechnology. A key limitation in such efforts concerns the availability of key co-factors, in this case NADPH, required for target pathways. Many of the strategies pursued for increasing NADPH availability in Escherichia coli involve manipulations to the central metabolism, which can create redox imbalances and overall growth defects. In this study we used a reactive oxygen species based selection to search for novel methods of increasing NADPH availability. We report a loss of function mutation in the gene hdfR appears to increase NADPH availability in E. coli. Additionally, we show this excess NADPH can be used to improve the production of 3HP in E. coli.


Subject(s)
Escherichia coli/physiology , Genetic Enhancement/methods , Lactic Acid/analogs & derivatives , Metabolic Engineering/methods , NADP/biosynthesis , Reactive Oxygen Species/metabolism , Biological Availability , Citric Acid Cycle/physiology , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Lactic Acid/isolation & purification , Lactic Acid/metabolism , Pentose Phosphate Pathway/physiology
12.
Genome Announc ; 5(5)2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28153899

ABSTRACT

We report here the draft genome sequences of two clinically isolated Acinetobacter baumannii strains. These samples were obtained from patients at the University of Colorado Hospital in 2007 and 2013 and encode an estimated 20 and 13 resistance genes, respectively.

13.
mSphere ; 2(1)2017.
Article in English | MEDLINE | ID: mdl-28217741

ABSTRACT

Antibiotic-resistant bacteria are an increasingly serious public health concern, as strains emerge that demonstrate resistance to almost all available treatments. One factor that contributes to the crisis is the adaptive ability of bacteria, which exhibit remarkable phenotypic and gene expression heterogeneity in order to gain a survival advantage in damaging environments. This high degree of variability in gene expression across biological populations makes it a challenging task to identify key regulators of bacterial adaptation. Here, we research the regulation of adaptive resistance by investigating transcriptome profiles of Escherichia coli upon adaptation to disparate toxins, including antibiotics and biofuels. We locate potential target genes via conventional gene expression analysis as well as using a new analysis technique examining differential gene expression variability. By investigating trends across the diverse adaptation conditions, we identify a focused set of genes with conserved behavior, including those involved in cell motility, metabolism, membrane structure, and transport, and several genes of unknown function. To validate the biological relevance of the observed changes, we synthetically perturb gene expression using clustered regularly interspaced short palindromic repeat (CRISPR)-dCas9. Manipulation of select genes in combination with antibiotic treatment promotes adaptive resistance as demonstrated by an increased degree of antibiotic tolerance and heterogeneity in MICs. We study the mechanisms by which identified genes influence adaptation and find that select differentially variable genes have the potential to impact metabolic rates, mutation rates, and motility. Overall, this work provides evidence for a complex nongenetic response, encompassing shifts in gene expression and gene expression variability, which underlies adaptive resistance. IMPORTANCE Even initially sensitive bacteria can rapidly thwart antibiotic treatment through stress response processes known as adaptive resistance. Adaptive resistance fosters transient tolerance increases and the emergence of mutations conferring heritable drug resistance. In order to extend the applicable lifetime of new antibiotics, we must seek to hinder the occurrence of bacterial adaptive resistance; however, the regulation of adaptation is difficult to identify due to immense heterogeneity emerging during evolution. This study specifically seeks to generate heterogeneity by adapting bacteria to different stresses and then examines gene expression trends across the disparate populations in order to pinpoint key genes and pathways associated with adaptive resistance. The targets identified here may eventually inform strategies for impeding adaptive resistance and prolonging the effectiveness of antibiotic treatment.

14.
ACS Synth Biol ; 6(1): 94-107, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27529436

ABSTRACT

The evolution of antibiotic resistance has engendered an impending global health crisis that necessitates a greater understanding of how resistance emerges. The impact of nongenetic factors and how they influence the evolution of resistance is a largely unexplored area of research. Here we present a novel application of CRISPR-Cas9 technology for investigating how gene expression governs the adaptive pathways available to bacteria during the evolution of resistance. We examine the impact of gene expression changes on bacterial adaptation by constructing a library of deactivated CRISPR-Cas9 synthetic devices to tune the expression of a set of stress-response genes in Escherichia coli. We show that artificially inducing perturbations in gene expression imparts significant synthetic control over fitness and growth during stress exposure. We present evidence that these impacts are reversible; strains with synthetically perturbed gene expression regained wild-type growth phenotypes upon stress removal, while maintaining divergent growth characteristics under stress. Furthermore, we demonstrate a prevailing trend toward negative epistatic interactions when multiple gene perturbations are combined simultaneously, thereby posing an intrinsic constraint on gene expression underlying adaptive trajectories. Together, these results emphasize how CRISPR-Cas9 can be employed to engineer gene expression changes that shape bacterial adaptation, and present a novel approach to synthetically control the evolution of antimicrobial resistance.


Subject(s)
CRISPR-Cas Systems , Escherichia coli/genetics , Drug Resistance, Bacterial/genetics , Epistasis, Genetic , Escherichia coli/growth & development , Evolution, Molecular , Gene Expression , Gene Targeting , Genes, Bacterial , Genetic Engineering , Stress, Physiological/genetics , Synthetic Biology
15.
ACS Synth Biol ; 5(12): 1566-1577, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27438180

ABSTRACT

The microbial ability to resist stressful environmental conditions and chemical inhibitors is of great industrial and medical interest. Much of the data related to mutation-based stress resistance, however, is scattered through the academic literature, making it difficult to apply systematic analyses to this wealth of information. To address this issue, we introduce the Resistome database: a literature-curated collection of Escherichia coli genotypes-phenotypes containing over 5,000 mutants that resist hundreds of compounds and environmental conditions. We use the Resistome to understand our current state of knowledge regarding resistance and to detect potential synergy or antagonism between resistance phenotypes. Our data set represents one of the most comprehensive collections of genomic data related to resistance currently available. Future development will focus on the construction of a combined genomic-transcriptomic-proteomic framework for understanding E. coli's resistance biology. The Resistome can be downloaded at https://bitbucket.org/jdwinkler/resistome_release/overview .


Subject(s)
Databases, Factual , Escherichia coli , Adaptation, Biological/genetics , Adaptation, Biological/physiology , Drug Resistance, Bacterial , Epistasis, Genetic , Escherichia coli/genetics , Escherichia coli/physiology , Genotype , Machine Learning , Mutation , Osmotic Pressure , Phenotype
16.
Genome Announc ; 4(3)2016 Jun 23.
Article in English | MEDLINE | ID: mdl-27340066

ABSTRACT

We report here the draft genome sequence of a multidrug-resistant Enterococcus faecalis strain, isolated from a patient at the University of Colorado Hospital. The genome assembly is 3,040,186 bp in length with 37.6% GC content. This isolate encodes eleven resistance genes, including those for glycopeptide, aminoglycoside, macrolide-lincosamide-streptogramin, and tetracycline resistance.

17.
Curr Opin Biotechnol ; 36: 107-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26319897

ABSTRACT

Metabolic engineers manipulate intricate biological networks to build efficient biological machines. The inherent complexity of this task, derived from the extensive and often unknown interconnectivity between and within these networks, often prevents researchers from achieving desired performance. Other fields have developed methods to tackle the issue of complexity for their unique subset of engineering problems, but to date, there has not been extensive and comprehensive examination of how metabolic engineers use existing tools to ameliorate this effect on their own research projects. In this review, we examine how complexity affects engineering at the protein, pathway, and genome levels within an organism, and the tools for handling these issues to achieve high-performing strain designs. Quantitative complexity metrics and their applications to metabolic engineering versus traditional engineering fields are also discussed. We conclude by predicting how metabolic engineering practices may advance in light of an explicit consideration of design complexity.


Subject(s)
Metabolic Engineering/methods , Genome , Humans , Metabolic Networks and Pathways , Proteins/genetics , Proteins/metabolism
18.
ACS Infect Dis ; 1(11): 555-67, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-27623410

ABSTRACT

The root cause of the antibiotic resistance crisis is the ability of bacteria to evolve resistance to a multitude of antibiotics and other environmental toxins. The regulation of adaptation is difficult to pinpoint due to extensive phenotypic heterogeneity arising during evolution. Here, we investigate the mechanisms underlying general bacterial adaptation by evolving wild-type Escherichia coli populations to dissimilar chemical toxins. We demonstrate the presence of extensive inter- and intrapopulation phenotypic heterogeneity across adapted populations in multiple traits, including minimum inhibitory concentration, growth rate, and lag time. To search for a common response across the heterogeneous adapted populations, we measured gene expression in three stress-response networks: the mar regulon, the general stress response, and the SOS response. While few genes were differentially expressed, clustering revealed that interpopulation gene expression variability in adapted populations was distinct from that of unadapted populations. Notably, we observed both increases and decreases in gene expression variability upon adaptation. Sequencing select genes revealed that the observed gene expression trends are not necessarily attributable to genetic changes. To further explore the connection between gene expression variability and adaptation, we propagated single-gene knockout and CRISPR (clustered regularly interspaced short palindromic repeats) interference strains and quantified impact on adaptation to antibiotics. We identified significant correlations that suggest genes with low expression variability have greater impact on adaptation. This study provides evidence that gene expression variability can be used as an indicator of bacterial adaptive resistance, even in the face of the pervasive phenotypic heterogeneity underlying adaptation.

19.
PLoS One ; 9(11): e113820, 2014.
Article in English | MEDLINE | ID: mdl-25422896

ABSTRACT

Advances in computational methods that allow for exploration of the combinatorial mutation space are needed to realize the potential of synthetic biology based strain engineering efforts. Here, we present Constrictor, a computational framework that uses flux balance analysis (FBA) to analyze inhibitory effects of genetic mutations on the performance of biochemical networks. Constrictor identifies engineering interventions by classifying the reactions in the metabolic model depending on the extent to which their flux must be decreased to achieve the overproduction target. The optimal inhibition of various reaction pathways is determined by restricting the flux through targeted reactions below the steady state levels of a baseline strain. Constrictor generates unique in silico strains, each representing an "expression state", or a combination of gene expression levels required to achieve the overproduction target. The Constrictor framework is demonstrated by studying overproduction of ethylene in Escherichia coli network models iAF1260 and iJO1366 through the addition of the heterologous ethylene-forming enzyme from Pseudomonas syringae. Targeting individual reactions as well as combinations of reactions reveals in silico mutants that are predicted to have as high as 25% greater theoretical ethylene yields than the baseline strain during simulated exponential growth. Altering the degree of restriction reveals a large distribution of ethylene yields, while analysis of the expression states that return lower yields provides insight into system bottlenecks. Finally, we demonstrate the ability of Constrictor to scan networks and provide targets for a range of possible products. Constrictor is an adaptable technique that can be used to generate and analyze disparate populations of in silico mutants, select gene expression levels and provide non-intuitive strategies for metabolic engineering.


Subject(s)
Computational Biology , Ethylenes/metabolism , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...