KEGG: lmf:LMOf2365_1849
Stp encodes a functional Mn²⁺-dependent serine-threonine phosphatase similar to PPM eukaryotic phosphatases (Mg²⁺- or Mn²⁺-dependent protein phosphatase) and is required for growth of L. monocytogenes in murine infection models . The phosphatase plays a crucial role in regulating the elongation factor EF-Tu, which is its first identified target. When an stp deletion mutation is introduced, L. monocytogenes exhibits decreased sensitivity to kirromycin, an antibiotic known to inhibit EF-Tu function . This altered antibiotic sensitivity suggests that Stp influences bacterial survival within the infected host through post-translational regulation mechanisms.
Research has demonstrated that signal transduction systems based on reversible phosphorylation are critical for L. monocytogenes to survive and grow in various host and environmental conditions. Among these regulatory elements, Stp stands out as one of only two putative serine-threonine phosphatases identified in the genome sequence .
Serotype 4b strains cause the majority of listeriosis clinical cases and outbreaks despite representing only one of 15 serovars within the genus . This clinical predominance occurs because:
Serotype 4b strains possess specific wall teichoic acid (WTA) glycosylation patterns that contribute to their virulence
Four major hypervirulent clonal complexes (CCs) - CC1, CC2, CC4, and CC6 - are predominantly found within serotype 4b
CC4 is significantly overrepresented among human isolates compared to food isolates, suggesting enhanced pathogenicity
Comparative studies have shown that serotype 4b strains account for a significantly higher proportion of clinical isolates (particularly in outbreaks) than would be expected based on their prevalence in food sources, with only 5.1% of clinical isolates carrying premature stop codon mutations compared to 45% of food isolates .
The primary verified target of Stp is the elongation factor EF-Tu . Post-translational phosphorylation of EF-Tu prevents:
Its binding to amino-acylated transfer RNA
Its binding to kirromycin (an antibiotic inhibitor of EF-Tu function)
This regulatory mechanism appears critical for bacterial survival during infection. While EF-Tu is the first identified target, the phosphatase likely has multiple substrates given the complexity of phosphorylation-based regulatory networks in bacterial pathogens. Ongoing phosphoproteomic studies are needed to identify additional substrates involved in various aspects of L. monocytogenes physiology and virulence.
For optimal expression and purification of recombinant Stp, researchers should consider:
Expression Systems:
E. coli BL21(DE3) with pET-based vectors for high-yield expression
Baculovirus-insect cell system for eukaryotic post-translational modifications when needed
Cell-free protein synthesis systems for proteins that may be toxic to host cells
Purification Protocol:
Transform expression vector containing the stp gene into appropriate host cells
Induce protein expression (typically with IPTG for E. coli systems)
Harvest cells and lyse using sonication or pressure-based methods
Perform initial purification using Ni-NTA affinity chromatography (for His-tagged constructs)
Apply secondary purification through ion exchange chromatography
Perform final polishing step using size-exclusion chromatography
Verify purity using SDS-PAGE and Western blotting
Critical Considerations:
Include Mn²⁺ in buffers (typically 1-5 mM) to maintain enzyme activity
Optimize temperature and pH (generally 4°C during purification; pH 7.0-7.5)
Include protease inhibitors to prevent degradation
Test activity immediately after purification using para-nitrophenyl phosphate (pNPP) assay
To generate stp deletion mutants in L. monocytogenes serotype 4b:
Allelic Exchange Method:
Construct a deletion vector containing approximately 1 kb of upstream and downstream flanking regions of the stp gene
Use a temperature-sensitive plasmid (e.g., pKSV7 or pIMK)
Transform the construct into L. monocytogenes serotype 4b
Select transformants under permissive conditions
Promote integration by shifting to non-permissive temperature with antibiotic selection
Release the integrated plasmid by growth without antibiotic at permissive temperature
Screen colonies for gene deletion using PCR
Verification Approaches:
PCR verification with primers flanking the deleted region
Whole genome sequencing to confirm deletion and absence of secondary mutations
Southern blot analysis to verify single genomic alterations
RT-PCR to confirm absence of stp transcript
Western blot to confirm absence of Stp protein
Phenotypic verification through kirromycin sensitivity assays
The deletion of stp in L. monocytogenes results in several observable phenotypes:
In vitro phenotypes:
Decreased sensitivity to kirromycin (an antibiotic that targets EF-Tu)
Altered phosphorylation status of EF-Tu
Potentially altered protein synthesis rates and fidelity
Changes in stress response mechanisms
In vivo phenotypes:
Decreased ability to grow and survive within host cells
Potentially altered interaction with host immune responses
Changes in tissue tropism or dissemination patterns
These phenotypic changes underscore the importance of Stp in regulating L. monocytogenes virulence and adaptation to the host environment through post-translational modifications of key proteins involved in bacterial physiology.
Recommended Methods for Studying Stp Activity:
Substrate-Based Assays:
pNPP Assay: Measures the release of para-nitrophenol from para-nitrophenyl phosphate
Advantages: Simple, colorimetric readout
Limitations: Non-specific substrate
Phosphopeptide-Based Assays:
Use synthetic peptides mimicking known Stp substrates like EF-Tu phosphorylation sites
Measure dephosphorylation using:
Malachite green assay (detects released phosphate)
Mass spectrometry (directly detects dephosphorylated peptides)
In-Gel Phosphatase Assays:
For analysis of native complexes and molecular weight determination
Incorporate radioactive substrates or fluorescent substrates
Kinetic Analysis:
Determine Km, Vmax, and kcat values using varying substrate concentrations
Test enzyme activity under different conditions:
pH range (typically 6.0-8.0)
Temperature range (25-42°C)
Divalent cation concentrations (Mn²⁺, Mg²⁺)
Potential inhibitors
Inhibitor Studies:
Test sensitivity to classical phosphatase inhibitors:
Okadaic acid
Calyculin A
Fluoride
Vanadate compounds
Identifying novel Stp substrates requires a multi-faceted approach:
Phosphoproteomic Approaches:
Comparative Phosphoproteomics:
Compare phosphorylation profiles between wild-type and Δstp mutants
Sample preparation: TCA precipitation followed by tryptic digestion
Phosphopeptide enrichment: TiO₂ or IMAC
LC-MS/MS analysis with label-free quantification or SILAC
In vitro Dephosphorylation Assays:
Incubate cell lysates with recombinant Stp
Identify dephosphorylated proteins by proteomics
Biochemical Approaches:
Affinity-Based Methods:
Use substrate-trapping mutants of Stp (e.g., D→A mutations in catalytic site)
Perform pull-down assays followed by mass spectrometry
Verify interactions using co-immunoprecipitation
Yeast Two-Hybrid Screening:
Use Stp as bait to identify interacting proteins
Confirm physical interactions using other methods
Bioinformatic Analysis:
Identify proteins with phosphorylation motifs similar to known Stp substrates
Focus on proteins involved in virulence, stress response, and cell wall functions
Prioritize candidates from serotype 4b-specific genetic regions
Validation Methods:
In vitro dephosphorylation assays with purified candidate proteins
Site-directed mutagenesis of putative phosphorylation sites
Phenotypic analysis of phosphomimetic and phosphoablative mutants
In Vitro Cellular Models:
Macrophage Infection Models:
Cell lines: J774, RAW264.7, THP-1
Primary mouse bone marrow-derived macrophages
Measurements: Bacterial survival, phagosomal escape, cytokine production
Epithelial Cell Invasion Assays:
Cell lines: Caco-2, HepG2, HeLa
Measurements: Adhesion, invasion, intracellular growth, cell-to-cell spread
Tissue Explant Models:
Placental tissue for maternal-fetal transmission studies
Brain tissue for blood-brain barrier crossing studies
In Vivo Animal Models:
Mouse Models:
Intravenous infection model (systemic listeriosis)
Oral infection model (natural route)
Galleria mellonella (wax moth larvae) as an alternative model
Specific Applications:
Model Selection Considerations:
| Model Type | Applications | Advantages | Limitations |
|---|---|---|---|
| Macrophage cell lines | Initial screening of virulence | Easy to manipulate, reproducible | Lack host complexity |
| Primary cells | Validation of cell line findings | More physiologically relevant | Donor variability |
| Mouse models | Systemic virulence assessment | Full immune response context | Ethical considerations, cost |
| Galleria mellonella | Rapid virulence screening | Ethical advantage, cost-effective | Limited immune system relevance |
| Tissue explants | Tissue-specific interactions | Maintains tissue architecture | Short experimental window |
When studying Stp specifically, researchers should consider models that emphasize intracellular growth phases and dissemination, as phosphorylation-based signaling likely plays key roles in adapting to changing host environments during infection.
While limited comparative data exists, researchers should consider:
Sequence Comparison Analysis:
Perform multiple sequence alignments of Stp from different serotypes
Focus on catalytic domains and regulatory regions
Identify serotype-specific variations that might affect function
Enzymatic Activity Comparison:
Express and purify Stp from multiple serotypes (1/2a, 1/2b, 4b)
Compare substrate specificity and kinetic parameters
Test activity under various environmental conditions
Cross-Complementation Studies:
Express serotype 4b Stp in Δstp mutants of other serotypes
Assess restoration of virulence phenotypes
Identify serotype-specific functional differences
Research indicates that serotype 4b strains generally exhibit enhanced virulence compared to other serotypes, with several hypervirulent clones (CC1, CC2, CC4, and CC6) predominantly found within this serotype . The functional differences in regulatory proteins like Stp may contribute to this enhanced virulence, making comparative studies particularly valuable.
Stp likely plays crucial roles in environmental adaptation through:
Stress Response Regulation:
Temperature fluctuations (growth at refrigeration temperatures)
pH changes (survival in acidic food products)
Osmotic stress (high salt environments)
Biofilm Formation:
Potential regulation of proteins involved in cell surface adhesion
Modulation of extracellular matrix components
Influence on bacterial communication systems
Antimicrobial Resistance:
Regulation of cell wall modifications
Modulation of efflux pump expression or activity
Altered susceptibility patterns to sanitizers and preservatives
Research approaches should include:
Transcriptomic comparison of wild-type and Δstp strains under various stresses
Biofilm formation assays under food-relevant conditions
Testing antimicrobial susceptibility profiles across environmental conditions
Targeting Stp or its regulatory pathways could lead to novel control strategies:
Inhibitor Development:
Design small molecule inhibitors specific to Stp active site
Target unique structural features not present in host phosphatases
Develop peptidomimetic inhibitors based on substrate recognition motifs
Anti-virulence Approaches:
Target downstream effectors of Stp-mediated regulation
Develop compounds that disrupt phosphorylation-dependent signaling
Consider combination approaches targeting multiple virulence mechanisms
Biocontrol Strategies:
Engineer bacteriophages expressing Stp inhibitors
Develop competitive exclusion approaches using attenuated strains
Create CRISPR-Cas delivery systems targeting stp gene
These approaches may be particularly effective against serotype 4b strains, which are responsible for the majority of human listeriosis cases despite being less prevalent in food than other serotypes .