This protein is a component of the PstSACB ABC transporter complex, responsible for phosphate import and energy coupling to the transport system.
KEGG: vvy:VVA0372
Phosphate import ATP-binding protein PstB 2 (pstB2) functions as a critical component of the phosphate-specific transport (Pst) system in Vibrio vulnificus, which is essential for bacterial phosphate homeostasis. While not directly characterized as a primary virulence factor like RtxA1 or VvhA (hemolysin), pstB2 plays an important role in bacterial survival within phosphate-limited environments such as human serum.
Research methodologically approaches this by:
Comparing growth rates of wild-type and pstB2-deficient strains in phosphate-limited media
Measuring phosphate uptake kinetics in various environmental conditions
Conducting survival assays in human serum to determine the protein's contribution to pathogenicity
Evidence suggests that phosphate acquisition systems contribute to bacterial fitness during infection, as demonstrated in serum-based survival studies identifying essential genes required for V. vulnificus growth in human serum .
The production of recombinant pstB2 typically employs a systematic expression protocol:
Gene cloning: The coding sequence for pstB2 is amplified from V. vulnificus genomic DNA (commonly strain CMCP6) using PCR with specific primers containing appropriate restriction sites.
Expression vector construction: The amplified gene is cloned into a prokaryotic expression vector (commonly pET-based systems) containing:
Inducible promoter (typically T7)
Affinity tag (6xHis-tag) for purification
Appropriate antibiotic resistance marker
Expression system: The recombinant vector is transformed into E. coli BL21(DE3) or similar expression strains.
Protein expression:
Culture growth at 37°C to mid-log phase (OD600 ≈ 0.6-0.8)
Induction with IPTG (typically 0.5-1.0 mM)
Post-induction growth at lower temperatures (16-25°C) for 4-16 hours
Purification:
Cell lysis (sonication or pressure-based disruption)
Immobilized metal affinity chromatography (IMAC)
Size exclusion chromatography for higher purity
The purified protein typically achieves >85% purity as assessed by SDS-PAGE , making it suitable for functional and structural studies.
Methodologically rigorous assessment of pstB2's contribution to serum survival requires multi-faceted approaches:
Genetic manipulation strategy:
Generation of clean deletion mutant (ΔpstB2) using allelic exchange
Complementation with wild-type gene on plasmid or chromosomally integrated
Construction of point mutants affecting key functional residues
Consider potential redundancy by creating double mutants with related genes
Serum survival assay protocol:
Pre-culture bacteria in phosphate-limited media to induce Pst system
Standardize inoculum to specific CFU (typically 10³-10⁵ CFU/mL)
Expose to human serum (both normal and heat-inactivated) at 37°C
Sample at defined timepoints (0, 1, 2, 4, 8h)
Determine viable counts on selective media
Calculate survival ratio compared to heat-inactivated controls
Complementary approaches:
Transcriptional profiling (RNA-seq) to identify compensatory mechanisms
Phosphate uptake kinetics using radiolabeled ³²P
Competition assays between wild-type and mutant strains
In vivo infection models (mouse) to validate serum survival findings
Sophisticated analysis requires integration of these data sets to distinguish between direct effects of pstB loss versus secondary metabolic consequences. Transposon insertion sequencing (TIS) studies in V. vulnificus have established experimental frameworks for such evaluations, identifying genes essential for serum survival .
Resolving contradictory findings requires systematic methodological approaches:
Standardized experimental conditions:
Use defined minimal media with controlled phosphate concentrations
Standardize growth phase for all comparisons
Maintain identical temperature, pH, and salinity conditions
Employ multiple biological and technical replicates
Comparative genomic and proteomic analyses:
Complete sequence alignment of pstB homologs across Vibrio species
Structural modeling to identify species-specific variations
Synteny analysis of pst operons to identify organizational differences
Phylogenetic analysis to correlate functional differences with evolutionary distance
Functional complementation experiments:
Cross-species gene swap experiments (replacing native pstB with homologs)
Domain swap constructs to identify functional regions responsible for differences
Heterologous expression systems to test activity under identical cellular backgrounds
Advanced biochemical characterization:
Compare kinetic parameters (Km, Vmax) for ATP hydrolysis
Assess phosphate transport efficiency using identical membrane vesicle preparations
Determine protein-protein interaction profiles with other Pst components
Research indicates that phosphate transport systems can differ in their regulation and functionality even between closely related bacteria. Genome-wide studies of V. vulnificus have identified distinct ecological adaptations , suggesting species-specific metabolic adaptations that may extend to phosphate acquisition systems.
Investigation of pstB2's interaction with virulence networks requires integrated methodological approaches:
Transcriptional profiling strategy:
RNA-seq comparing wild-type and ΔpstB2 mutants under varying phosphate conditions
ChIP-seq to identify transcription factors responding to pstB2-dependent signals
Promoter-reporter fusion assays to validate transcriptional responses
Regulatory network identification:
Two-component system analysis (particularly PhoR-PhoB)
Quantification of cyclic-AMP levels and CRP-dependent gene expression
Analysis of iron-responsive regulators (Fur) that intersect with phosphate regulation
Assessment of quorum sensing systems (LuxO, SmcR) known to regulate virulence
Virulence factor expression measurement:
qRT-PCR quantification of key virulence genes (rtxA1, vvhA)
Western blot analysis of toxin production
Enzymatic activity assays for secreted virulence factors
Immunoblotting for capsular polysaccharide production
Evidence indicates complex regulatory networks in V. vulnificus that integrate multiple environmental signals. For example, CRP activates vvhBA transcription under glucose starvation , while iron can repress vvhA transcription via Fur . The status of phosphate acquisition likely integrates into these networks, with the PhoR-PhoB system potentially serving as a link between phosphate limitation and virulence regulation.
A comprehensive research methodology for investigating pstB2's role in biofilm formation includes:
Biofilm formation assay workflow:
Static microtiter plate assays with crystal violet staining
Flow cell systems with confocal microscopy for structure analysis
Quantification of extracellular polymeric substances (EPS)
Comparison between wild-type, ΔpstB2, and complemented strains
Testing under varying phosphate concentrations (0.1-2mM)
Environmental persistence evaluation:
Survival in artificial seawater with controlled nutrient levels
Microcosm experiments simulating natural environmental conditions
Competition assays with environmental isolates
Long-term starvation survival assessment
Molecular basis investigation:
Transcriptional profiling of biofilm-associated genes
Quantification of second messengers (c-di-GMP, (p)ppGpp)
Analysis of extracellular DNA and polysaccharide composition
Examination of cell surface hydrophobicity and adhesion properties
Research on V. vulnificus virulence factors suggests links between metabolic systems and biofilm formation. For instance, flagella components (flgK, flgL, flgE) identified in co-adaptation networks have been shown to affect biofilm formation . As phosphate limitation typically triggers biofilm formation in many bacteria, pstB2 likely plays a role in this environmental adaptation process.
An optimal knockout strategy requires careful genetic manipulation approaches:
In-frame deletion method:
Amplify ~1kb flanking regions upstream and downstream of pstB2
Fuse fragments with overlap extension PCR to maintain reading frame
Clone into suicide vector (e.g., pDM4 with sacB for counter-selection)
Perform two-step allelic exchange via homologous recombination
Screen for sucrose resistance and antibiotic sensitivity
Confirm deletion by PCR and sequencing of the junction region
Marker-less deletion considerations:
Utilize FLP/FRT or Cre/loxP recombination systems
Design junction points that maintain translational coupling
Preserve ribosome binding sites for downstream genes
Avoid disrupting operon regulatory elements
Validation of non-polar effects:
RT-PCR analysis of adjacent gene expression
Complementation with pstB2 alone to verify phenotype restoration
Transcriptional profiling to assess global effects
Growth rate comparison in various media
Alternative approaches:
Site-directed mutagenesis of key functional residues
CRISPR/Cas9-based genome editing for precise modifications
Conditional knockdown using antisense RNA or inducible promoters
Studies on V. vulnificus virulence genes have successfully employed clean deletion strategies to avoid polar effects. For example, research on capsule biogenesis genes demonstrated the importance of clean deletions when examining closely linked genes such as VV0357 and VV0358 .
Robust experimental designs for stress response comparison require:
Standardized growth protocol:
Define pre-culture conditions (medium, growth phase, temperature)
Normalize cell density across all experiments (OD600 ≈ 0.5)
Wash cells to remove residual media components
Use balanced experimental design with appropriate controls
Stress exposure matrix:
| Stress Condition | Variables to Test | Measurement Parameters | Timepoints |
|---|---|---|---|
| Phosphate limitation | 0, 0.1, 0.5, 1.0 mM Pi | Growth rate, survival | 0, 2, 4, 8, 24h |
| Oxidative stress | 0.1-5 mM H₂O₂ | Survival, enzyme activity | 0, 30, 60, 120min |
| Serum exposure | 10-100% human serum | CFU recovery, gene expression | 0, 1, 2, 4h |
| pH stress | pH 5.0-9.0 | Growth, metabolic activity | 0, 2, 4, 8h |
| Antimicrobial peptides | 1-100 μg/mL | MIC, membrane integrity | 0, 2, 4, 8h |
Multiparameter analysis:
Viability assessment (plate counts, live/dead staining)
Metabolic activity (ATP levels, resazurin reduction)
Membrane integrity (propidium iodide uptake)
Specific stress responses (ROS levels, pH homeostasis)
Statistical approach:
Minimum of three biological replicates
Two-way ANOVA to assess strain × condition interactions
Appropriate post-hoc tests with correction for multiple comparisons
Effect size calculation to quantify biological significance
This approach allows for systematic identification of conditions where pstB2 contributes to stress resilience, providing insights into its role in V. vulnificus pathogenicity. Similar experimental frameworks have been used in studies identifying virulence-associated genes in V. vulnificus .
Detection of phosphate transport requires sensitive methodological approaches:
Radioisotope-based transport assays:
Use ³²P-labeled inorganic phosphate (³²Pi)
Prepare inside-out membrane vesicles from expressing cells
Initiate transport with ATP addition
Sample at short intervals (15, 30, 60, 120 seconds)
Rapid filtration through nitrocellulose membranes
Scintillation counting for accurate quantification
Calculate initial rates from linear phase
Fluorescent phosphate analog approaches:
BCECF-based intracellular pH monitoring (proton-coupled transport)
Phosphate analogs with fluorescent properties
Real-time continuous monitoring using plate reader
Kinetic analysis for Km and Vmax determination
Indirect coupling assays:
ATPase activity measurement (colorimetric phosphate release)
Coupled enzyme assays (ATP regeneration systems)
Membrane potential sensitive dyes
Reconstitution system optimization:
Proteoliposome preparation with defined lipid composition
Co-reconstitution with complete Pst complex components
Varying ATP concentrations for kinetic parameter determination
Assessment of substrate specificity (Pi, arsenate, phosphite)
Data analysis considerations:
Correction for passive diffusion
Controlling for vesicle integrity and size
Protein quantification for specific activity calculation
Comparison with known phosphate transporters as references
These methodologies provide comprehensive functional assessment, distinguishing between ATP binding, hydrolysis, and actual phosphate translocation activities of pstB2. Similar approaches have been employed for characterizing transport systems in V. vulnificus and related bacteria .
Interpretation of expression differences requires rigorous analytical approaches:
Expression quantification methodology:
qRT-PCR with validated reference genes specific for V. vulnificus
RNA-seq with appropriate normalization (TPM, RPKM)
Western blot using specific antibodies against pstB2
Standardize growth conditions and sampling points
Statistical analysis framework:
Minimum sample size calculation based on expected effect size
Non-parametric tests for non-normally distributed data
Multiple testing correction (Benjamini-Hochberg FDR)
Correlation analysis with other genetic markers
Contextual interpretation guidelines:
Relate to phylogenetic grouping of isolates
Consider ecological niches and isolation sources
Analyze in context of other phosphate metabolism genes
Compare with established virulence factor expression patterns
Validation approaches:
Reporter gene fusions to confirm transcriptional differences
Complementation experiments across isolate backgrounds
Functional assays to correlate expression with phosphate uptake capacity
Research on V. vulnificus isolates has demonstrated significant genetic diversity with clinical implications. Genome-wide studies have identified distinct phylogenetic groups that correlate with virulence potential . Phosphate acquisition mechanisms may follow similar patterns, with analysis requiring consideration of the genetic background and ecological adaptation of each isolate.
Distinguishing direct from indirect effects requires integrated analytical frameworks:
Metabolomic profiling strategy:
Untargeted LC-MS/MS metabolomics of wild-type vs. mutant
Stable isotope labeling to track phosphate incorporation
Pathway enrichment analysis to identify affected metabolic modules
Temporal sampling to distinguish primary from secondary effects
Multi-omics integration approach:
Paired transcriptomic and proteomic analysis
Computational modeling of metabolic networks
Flux balance analysis to predict metabolic rerouting
Network analysis to identify regulatory hubs
Genetic interaction mapping:
Synthetic genetic array analysis with pstB2 deletion
Chemical genetic profiling under varying phosphate conditions
Suppressor mutation screening to identify compensatory pathways
Direct vs. indirect effect criteria:
| Effect Type | Temporal Pattern | Genetic Rescue | Metabolic Signature |
|---|---|---|---|
| Direct | Immediate | Only by pstB2 | Specific to Pi metabolism |
| Indirect | Delayed | Multiple genes | Broad metabolic changes |
| Compensatory | Adaptive | New equilibrium | Alternative pathway activation |
Statistical approaches:
Principal component analysis to separate direct vs. indirect effects
Time series analysis to establish causality
Bayesian network modeling for probabilistic relationships
This framework provides a systematic approach to deconvolute the complex effects of pstB2 disruption. Similar approaches have been employed in genome-wide epistasis studies (GWES) of V. vulnificus to identify co-adaptation networks and distinguish between direct genetic interactions and indirect effects .
Structure-based drug design approaches for pstB2 inhibitors require:
Structural characterization methodology:
X-ray crystallography of pstB2 in multiple conformational states
Cryo-EM of complete Pst transport complex
Molecular dynamics simulations to identify cryptic binding sites
Hydrogen-deuterium exchange mass spectrometry for conformational dynamics
Target site identification:
ATP binding pocket for competitive inhibitors
Interface regions between pstB2 and membrane components
Allosteric sites that prevent conformational cycling
Species-specific structural features absent in human transporters
Inhibitor design strategy:
Virtual screening of compound libraries against identified pockets
Fragment-based approach starting with ATP-competitive scaffolds
Structure-activity relationship optimization
Incorporation of physico-chemical properties for penetration
Validation cascade:
In vitro ATPase inhibition assays
Transport inhibition in membrane vesicles
Bacterial growth inhibition under phosphate limitation
Toxicity assessment against human cell lines
In vivo efficacy in infection models
Research on V. vulnificus has identified several potential therapeutic targets, including the RtxA1 toxin , which has been successfully targeted for vaccine development. Structural insights into essential transport systems like pstB2 could similarly inform therapeutic development, particularly given the increasing antibiotic resistance observed in V. vulnificus .
Assessment of pstB2 as a therapeutic target requires systematic in vivo evaluation:
Animal model selection and design:
Iron-overloaded mouse model (mimics human risk factors)
Wound infection model with controlled bacterial inoculum
Gastrointestinal infection model for food-borne route
Use of both wild-type and pstB2-deficient strains with bioluminescent reporters
Virulence parameter measurement:
Bacterial burden in blood and tissues (spleen, liver)
Survival curve analysis with Kaplan-Meier statistics
Cytokine profiling to assess inflammatory response
Histopathological examination of infected tissues
Inhibitor evaluation protocol:
Pharmacokinetic studies to establish dosing regimen
Prophylactic vs. therapeutic administration timing
Combination with standard antibiotics
Different administration routes (oral, intraperitoneal, topical)
Advanced infection modeling:
Humanized mouse models with human serum components
Ex vivo human tissue infection models
Microfluidic organ-on-chip systems for controlled infection studies
Data from previous virulence studies in V. vulnificus demonstrate that disruption of essential transport systems can significantly attenuate virulence. For example, capsule biosynthesis mutants show dramatically reduced virulence in mouse models , and similar effects might be expected for phosphate transport deficiencies under the phosphate-limited conditions encountered during infection.
Investigation of pstB2 genetic diversity requires clinical correlation approaches:
Genetic diversity assessment:
Whole genome sequencing of clinical isolates
SNP and structural variant identification in pstB2
Haplotype analysis and phylogenetic grouping
Functional prediction of amino acid substitutions
Clinical correlation methodology:
Retrospective analysis of patient outcomes linked to isolate genetics
Case-control studies comparing severe vs. mild infections
Standardized clinical data collection with severity scores
Multivariate analysis controlling for host factors
Functional validation:
Site-directed mutagenesis to recreate clinical variants
Transport activity assays comparing variant proteins
Competition assays between variant strains in serum
Transcriptional response profiling of variant strains
Predictive model development:
Machine learning approaches integrating genetic and clinical data
Risk stratification based on bacterial and host factors
Validation in prospective clinical cohorts
Florida Department of Health surveillance data shows significant variability in clinical outcomes of V. vulnificus infections, with mortality rates of approximately 20-25% . Genetic determinants of virulence, including potential variations in essential systems like phosphate transport, may contribute to this clinical heterogeneity.
Analysis of pstB2 regulation under antibiotic stress requires:
Expression analysis methodology:
qRT-PCR of pstB2 under sub-inhibitory antibiotic concentrations
Reporter fusions (pstB2-promoter:GFP) for real-time monitoring
Western blot to confirm protein-level changes
Single-cell analysis to detect population heterogeneity
Antibiotic exposure protocol:
Test multiple antibiotic classes (β-lactams, aminoglycosides, tetracyclines)
Dose-response assessment (0.1x-1x MIC)
Time-course analysis (immediate vs. adaptive responses)
Combinatorial antibiotic testing
Functional consequence evaluation:
Phosphate uptake measurement under antibiotic stress
Bacterial survival in serum following antibiotic exposure
Metabolic activity assessment (ATP levels, respiration)
Membrane potential and permeability changes
Clinical implication assessment:
Antibiotic efficacy testing in phosphate-limited conditions
Persister cell formation associated with phosphate transport
Sequential treatment protocols based on metabolic adaptations
Host-relevant microenvironments (varying phosphate availability)
Research indicates that V. vulnificus has begun developing resistance to certain antibiotics , and understanding how essential systems like phosphate transport respond to antibiotic stress could inform improved treatment strategies. Stress responses often engage multiple regulatory networks, and phosphate acquisition systems might be upregulated as part of general stress adaptation mechanisms.
Investigation of serum effects on pstB2 requires systematic methodological approaches:
Expression analysis in serum:
Transcriptional profiling (RNA-seq) comparing growth in serum vs. control media
qRT-PCR targeting the complete pst operon
Western blotting to confirm protein-level changes
Reporter gene constructs for real-time monitoring
Functional activity assessment:
Radiolabeled phosphate uptake assays in serum-exposed bacteria
Competition assays with ΔpstB2 mutants in normal vs. heat-inactivated serum
Phosphate depletion measurement from growth medium
ATP hydrolysis assays with membrane preparations
Serum factor identification:
Fractionation of serum components
Testing purified serum proteins (transferrin, albumin)
Assessment of complement effects
Iron chelation experiments to distinguish from iron-limitation responses
Physiological relevance determination:
Comparison of different host species sera
Correlation with phosphate levels in patient samples
Ex vivo infection models with human blood
Link to disease progression markers