KEGG: sas:SAS1019
The phosphoenolpyruvate-protein phosphotransferase system (PTS) in S. aureus is a major mechanism for carbohydrate uptake and phosphorylation. It operates through a series of phosphoryl transfers starting from phosphoenolpyruvate (PEP) as the phosphoryl donor. The system involves three essential catalytic entities: Enzyme I, Enzyme II (with various sugar-specific variants), and HPr (heat-stable, histidine-phosphorylatable protein) .
This system is characterized by four successive phosphoryl transfers:
Initial autophosphorylation of Enzyme I using PEP as substrate
Transfer of the phosphoryl group from Enzyme I to HPr
Transfer from HPr to the sugar-specific EIIA component
Final transfer to the sugar via EIIB during transport through the membrane-bound EIIC
Unlike other transport systems, the PTS simultaneously transports and phosphorylates carbohydrates, coupling these processes to conserve energy .
SAS1019 refers to a partial recombinant form of the phosphoenolpyruvate-protein phosphotransferase from S. aureus. Based on general PTS research, this protein likely represents Enzyme I (EI), the first component in the PTS phosphorylation cascade. The molecular weight of Enzyme I in S. aureus has been estimated at approximately 100,000 ± 15% Da .
Functionally, if SAS1019 is indeed Enzyme I, it catalyzes the initial phosphorylation step in the PTS cascade, using PEP to autophosphorylate at a conserved histidine residue. This phosphoryl group is then transferred to the HPr protein, initiating the cascade that ultimately results in sugar uptake and phosphorylation .
The PTS system serves several critical functions in S. aureus:
Primary metabolic functions:
Regulatory functions:
Controls carbon catabolite repression
Regulates expression of genes involved in central carbon metabolism
Coordinates carbon and nitrogen metabolism
Connection to virulence:
While specific information on S. aureus is limited in the search results, studies in related bacteria suggest PTS components can influence virulence. For example, in Borrelia burgdorferi, mutations in ptsG (encoding EIIBC components) eliminated mouse infectivity . The PTS is likely involved in S. aureus adaptation to host environments through regulation of metabolism and gene expression.
Identity verification:
SDS-PAGE to confirm molecular weight (~100,000 Da for EI components)
Western blotting with anti-His or anti-PTS component antibodies
Mass spectrometry (MALDI-TOF or LC-MS/MS) for accurate mass determination and peptide mapping
N-terminal sequencing to confirm protein identity
Activity assays:
Colorimetric assays based on formation of o-nitrophenyl-β-d-galactoside-6-phosphate and its hydrolysis by 6-phospho-β-galactosidase
Coupled enzyme assays measuring pyruvate production from PEP
Direct measurement of phosphoryl transfer using:
In vitro reconstitution of the complete PTS cascade with purified components
Research on PTS mutations has revealed significant impacts on bacterial physiology:
Metabolic effects:
Accumulation of metabolic intermediates (e.g., MurNAc-GlcNAc in MurP mutants)
Altered carbon catabolite repression patterns
Changes in central carbon metabolism flux
Growth phenotypes:
Mutants in mupG and murQ components show slight growth defects during late exponential and stationary phases
Sugar utilization profiles are altered in PTS component mutants
Virulence implications:
While specific S. aureus data is limited, research in related bacteria is informative:
In Borrelia burgdorferi, ptsG mutants lost infectivity in mice but survived normally in ticks
PTS mutations can affect adaptation to host environments through altered metabolism
Genetic compensation:
Transcriptome analysis reveals compensatory changes in gene expression following PTS mutations
PTS mutations trigger expression changes in metabolic and regulatory genes
The PTS system functions as a sophisticated signaling network that integrates multiple metabolic inputs:
Integration with carbon metabolism:
Senses the PEP:pyruvate ratio, reflecting the energy state of the cell
Responds to glycerol, pyruvate, oxaloacetate, and serine through distinct mechanisms
Coordinates with glycolysis, TCA cycle, and gluconeogenesis
Cross-talk with nitrogen metabolism:
In some bacteria, α-ketoglutarate (which accumulates under nitrogen limitation) affects PTS activity, though this interaction varies between species
PTS components regulate nitrogen-related gene expression in some bacteria
Signaling network properties:
This integration allows the PTS to function as a global nutritional sensor rather than simply a sugar transport system.
In vitro interaction studies:
Pull-down assays with purified components
Surface plasmon resonance (SPR) to measure binding kinetics
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Chemical cross-linking followed by mass spectrometry
In vitro reconstitution of phosphoryl transfer using purified components
In vivo interaction studies:
FRET analysis with fluorescent protein fusions:
Bacterial two-hybrid systems
Co-immunoprecipitation from cell lysates
Split-reporter assays (luciferase, GFP)
Key findings from FRET studies:
Cytoplasmic PTS components are recruited to membrane transporters upon sugar stimulation
Interactions between different PTS proteins respond similarly to all PTS sugars
The relative response amplitudes upon stimulation with saturating concentrations of different PTS sugars are equal across different protein pairs
These techniques have revealed that the phosphorylation state of proteins within the network can rapidly equilibrate, suggesting reversibility is a general property of the PTS.
Key residues for targeted mutagenesis:
Conserved histidine phosphorylation sites
PEP binding pocket residues
Protein-protein interaction interfaces
Regulatory domains
Mutagenesis strategy:
Identify conserved residues through sequence alignment of PTS proteins across species
Design mutagenesis primers introducing substitutions:
Histidine → Alanine (eliminates phosphorylation)
Histidine → Glutamate (mimics phosphorylation)
Conservative substitutions to assess specific chemical properties
Expression and analysis workflow:
Create mutant constructs using PCR-based methods (QuikChange, overlap extension)
Express wild-type and mutant proteins under identical conditions
Purify to homogeneity using affinity tags and chromatography
Compare structural integrity using circular dichroism or thermal shift assays
Assess functional impacts through:
In vitro phosphoryl transfer assays
Protein-protein interaction studies
Complementation of PTS-deficient bacterial strains
Validation approaches:
Complementation studies in S. aureus PTS mutants
In vivo assessment of sugar uptake and metabolism
Structural analysis of mutant proteins by X-ray crystallography or NMR
Radioactive labeling approaches:
[³²P]PEP as phosphoryl donor:
Follow phosphoryl transfer through sequential PTS components
Detect radiolabeled intermediates by SDS-PAGE and autoradiography
Time-course experiments to determine transfer kinetics
Quench-flow experiments for rapid kinetics
[¹⁴C] or [³H]-labeled sugars:
Monitor uptake and phosphorylation in whole cells
Measure accumulation of labeled sugar phosphates
Compare wild-type versus mutant strains
Stable isotope applications:
¹³C-labeled sugars or PEP:
Track carbon flow through metabolic pathways
Identify branch points and flux distributions
Combine with mass spectrometry for metabolite analysis
¹⁸O-labeled phosphate:
Distinguish newly formed phosphoryl bonds
Follow phosphate movement through the system
Mass spectrometry analysis of phosphorylated intermediates
NMR applications:
Real-time monitoring of phosphoryl transfer reactions
Structural analysis of phosphorylated intermediates
Investigation of protein conformational changes
Experimental workflow:
Prepare isotope-labeled substrates or proteins
Initiate phosphoryl transfer reaction
Sample at defined timepoints
Separate components by gel electrophoresis, HPLC, or other methods
Detect isotope incorporation by appropriate methods
Analyze kinetics and pathway intermediates
Recent systems biology studies have transformed our understanding of the PTS from a simple sugar transport system to a sophisticated sensory and regulatory network:
Network sensing properties:
Integration of multiple inputs:
Beyond PTS sugars, the system responds to metabolites like glycerol, pyruvate, oxaloacetate, and serine
These signals are propagated through the network in different ways:
Global regulation:
Transcriptome analysis reveals extensive regulation beyond sugar metabolism
PTS components modulate expression of genes involved in virulence, stress response, and other functions
Mathematical modeling insights:
Kinetic models predict optimal regulatory strategies based on carbon source quality
Default sugar uptake through uninduced PTS correlates with carbon source quality
CRISPR-based approaches:
CRISPR interference (CRISPRi) for tunable repression of PTS components
CRISPR activation (CRISPRa) for overexpression studies
CRISPR-Cas9 genome editing for precise chromosomal modifications
Advanced imaging technologies:
Super-resolution microscopy to visualize PTS component localization
Single-molecule FRET to observe individual phosphoryl transfer events
Live-cell imaging with fluorescent sensors for sugar uptake and phosphorylation
Computational approaches:
Molecular dynamics simulations of PTS component interactions
Systems-level modeling of PTS regulatory networks
Machine learning analysis of PTS-dependent gene expression patterns
High-throughput methodologies:
Multiplexed assays for PTS activity across multiple conditions
Deep mutational scanning to comprehensively map functional residues
Automated microfluidic systems for kinetic measurements
Synthetic biology applications:
Engineering PTS components for non-native sugar utilization
Creating biosensors based on PTS regulatory mechanisms
Developing metabolic switches controlled by PTS signaling
PTS research offers several promising avenues for antimicrobial development:
Targeting PTS components:
Inhibitors of Enzyme I or HPr could disrupt carbon metabolism broadly
Sugar-specific EII inhibitors might prevent utilization of specific carbon sources
Allosteric modulators of PTS protein interactions could disrupt signaling
Metabolic vulnerabilities:
PTS mutant studies reveal metabolic bottlenecks that could be targeted
Understanding carbon source utilization in infection environments identifies crucial pathways
PTS-dependent gene regulation may reveal non-obvious drug targets
Connection to virulence:
In some bacteria like B. burgdorferi, PTS components (PtsG) are essential for infectivity
PTS regulation affects expression of virulence factors in some pathogens
Targeting PTS could attenuate virulence without necessarily killing bacteria (anti-virulence approach)
Potential advantages:
Novel targets outside traditional antibiotic classes
Potential for pathogen-specific targeting based on PTS differences
Metabolic targets may be less susceptible to conventional resistance mechanisms
Challenges:
Redundant carbon utilization pathways may bypass PTS inhibition
Human glucose transporters could be inadvertently affected
Resistance might develop through mutations in target sites or expression of alternative transporters
The PTS system plays multiple roles in S. aureus pathogenesis across different infection environments:
Blood and tissue infections:
PTS components facilitate adaptation to glucose-limited environments
Sugar sensing via PTS may regulate expression of immune evasion factors
Biofilm formation:
PTS-dependent carbon metabolism influences biofilm development
Altered metabolism in biofilms may rely on alternative PTS substrates
Cell wall recycling mediated by MurP and MupG affects peptidoglycan remodeling in biofilms
Intracellular survival:
Experimental evolution of S. aureus in macrophages produces mutations in metabolic genes
Small colony variants (SCVs) with altered metabolism show enhanced intracellular survival
These variants exhibit increased survival in macrophages and human blood, along with vancomycin resistance
Host-specific adaptation:
PTS system helps sense available nutrients in different host niches
Functions as a global nutrient sensor rather than detecting specific sugars
Coordinates metabolic adaptation to changing host environments
Research in other pathogens supports these roles - for example, in Borrelia burgdorferi, the PtsG component is essential for establishing mouse infection but not for survival in ticks .
PTS proteins contribute to antibiotic resistance through several mechanisms:
Metabolic adaptation:
Altered carbon metabolism affects susceptibility to antibiotics targeting active growth
PTS regulation influences cell wall synthesis and turnover pathways
The recovery of peptidoglycan turnover products via MurP affects cell wall homeostasis
Regulatory connections:
PTS-dependent gene regulation affects expression of resistance determinants
Metabolic state sensed by PTS influences antibiotic tolerance
Small colony variants (SCVs) with altered metabolism show increased resistance to vancomycin
Cell wall metabolism:
The PTS component MurP is involved in recovery of the cell wall turnover product MurNAc-GlcNAc
MupG hydrolyzes MurNAc 6-phosphate-GlcNAc intracellularly
These processes affect peptidoglycan recycling and potentially antibiotic susceptibility
Stress response coordination:
PTS functions as a sensor for nutrient availability
This sensing capability helps coordinate stress responses that contribute to antibiotic tolerance
Metabolic adaptations sensed through PTS may trigger persistence mechanisms
Experimental evolution studies have shown that S. aureus adaptation to intracellular environments can produce variants with both enhanced survival in macrophages and increased antibiotic resistance .
Structural analysis tools:
Homology modeling servers (SWISS-MODEL, I-TASSER, Phyre2)
Molecular dynamics simulations (GROMACS, AMBER, NAMD)
Protein-protein docking (HADDOCK, ClusPro, ZDOCK)
Binding site prediction (SiteMap, FTMap, CASTp)
Molecular visualization (PyMOL, Chimera, VMD)
Sequence analysis tools:
Multiple sequence alignment (Clustal Omega, MUSCLE, T-Coffee)
Conservation analysis (ConSurf, SIFT, PolyPhen)
Domain prediction (SMART, Pfam, InterPro)
Phosphorylation site prediction (NetPhos, GPS, PPSP)
Transmembrane topology prediction (TMHMM, Phobius)
Systems biology approaches:
Network analysis (Cytoscape, STRING, InnateDB)
Metabolic modeling (COBRA Toolbox, PathwayTools)
Gene expression analysis (DESeq2, EdgeR, GSEA)
Kinetic modeling (COPASI, CellDesigner)
Flux balance analysis (BiGG, COBRA)
Application examples:
Homology modeling of S. aureus PTS components based on solved structures
Molecular dynamics simulations of phosphoryl transfer mechanisms
Systems models of PTS regulation integrating transcriptomic data
Prediction of critical residues for mutagenesis studies
Virtual screening for potential PTS inhibitors
Network analysis approaches uncover system-level properties of the PTS:
Network representation approaches:
Protein-protein interaction networks:
Nodes represent PTS components
Edges represent physical interactions or phosphoryl transfer
Weighted by interaction strength or kinetic parameters
Regulatory networks:
Include PTS-regulated genes and proteins
Capture transcriptional and post-translational regulation
Represent feedback loops and cross-regulation
Metabolic networks:
Integrate PTS with broader metabolic pathways
Capture substrate flows and energy coupling
Predict metabolic responses to perturbations
Emergent properties revealed:
Network analysis techniques:
Topological analysis to identify key nodes and bottlenecks
Dynamic analysis to predict system responses to perturbations
Constraint-based analysis to predict feasible metabolic states
Comparative analysis across species to identify evolutionary patterns
Several promising research directions emerge from current knowledge:
Structural biology:
Determining high-resolution structures of complete S. aureus PTS complexes
Characterizing dynamic conformational changes during phosphoryl transfer
Mapping interaction interfaces between PTS components and regulatory targets
Structural basis for sugar specificity in different EII transporters
Systems biology:
Comprehensive mapping of the PTS interactome in S. aureus
Integration of transcriptomic, proteomic, and metabolomic responses to PTS perturbations
Development of predictive models for PTS regulation during infection
Comparative analysis of PTS networks across S. aureus strains with different virulence properties
Host-pathogen interactions:
Role of PTS in sensing host-specific nutritional environments
Influence of PTS on immune evasion mechanisms
Contribution to formation of antibiotic-tolerant persister cells
Role in biofilm development and chronic infection
Therapeutic applications:
High-throughput screening for inhibitors of key PTS components
Development of PTS-targeted antimicrobial peptides
Exploration of PTS components as vaccine targets
Design of metabolic adjuvants to enhance antibiotic efficacy
Technological innovations:
Development of real-time sensors for PTS activity in living cells
Application of synthetic biology approaches to reprogram PTS regulation
Creation of cellular diagnostics based on PTS function
Single-cell analysis of PTS activity during infection
Interdisciplinary approaches offer powerful ways to advance PTS research:
Integrative structural biology:
Combining X-ray crystallography, cryo-EM, and NMR spectroscopy
Integrating computational modeling with experimental data
Using hydrogen-deuterium exchange mass spectrometry (HDX-MS)
Applying single-molecule techniques to observe dynamic processes
Synthetic biology:
Engineering minimal PTS systems with defined components
Creating biosensors based on PTS signaling mechanisms
Developing orthogonal PTS pathways for biotechnology applications
Rewiring PTS regulation to create new cellular behaviors
Advanced imaging:
Super-resolution microscopy to visualize PTS component localization
Live-cell imaging with fluorescent biosensors
Correlative light and electron microscopy
Label-free imaging techniques (Raman microscopy, FTIR)
Machine learning applications:
Prediction of PTS component interactions
Analysis of complex phenotypic data from PTS mutants
Virtual screening for PTS inhibitors
Modeling evolutionary trajectories of PTS components
Host-pathogen systems biology:
Dual RNA-seq of infected host cells
Metabolic modeling of host-pathogen nutrient competition
Multi-omics integration across infection stages
Agent-based modeling of infection dynamics
These interdisciplinary approaches can reveal new insights about how the PTS functions as a global sensor of sugar influx and integrator of multiple metabolic signals, rather than just a sugar transport system .