Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes.
Note: All proteins are shipped with standard blue ice packs unless dry ice is specifically requested. Advance notice is required for dry ice shipping, and additional fees will apply.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
This protein is involved in the biosynthesis of isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP), essential building blocks of isoprenoid compounds. It catalyzes the conversion of 4-diphosphocytidyl-2-C-methyl-D-erythritol 2-phosphate (CDP-ME2P) to 2-C-methyl-D-erythritol 2,4-cyclodiphosphate (ME-CPP), releasing cytidine 5-monophosphate (CMP) in the process.
KEGG: vvy:VV2814
IspF (2-C-methyl-D-erythritol 2,4-cyclodiphosphate synthase) catalyzes a critical cyclization reaction in the MEP (methylerythritol phosphate) pathway, converting 4-diphosphocytidyl-2-C-methyl-D-erythritol 2-phosphate to 2-C-methyl-D-erythritol 2,4-cyclodiphosphate and CMP. This pathway is essential for isoprenoid biosynthesis in many bacteria, including V. vulnificus. The MEP pathway is particularly important during infection, as V. vulnificus experiences dramatic environmental changes moving from estuarine environments through the host's gastrointestinal tract and eventually into the bloodstream. These transitions require metabolic adaptations supported by isoprenoid-derived molecules essential for membrane integrity and various cellular functions .
The MEP pathway represents an attractive target for antimicrobial development because humans utilize the distinct mevalonate pathway for isoprenoid synthesis. Recent studies have demonstrated that synergistic approaches combining MEP pathway engineering with other metabolic pathways can significantly enhance production of valuable downstream products like isoprene, suggesting the pathway's critical metabolic role .
V. vulnificus IspF shares the conserved trimeric structure typical of IspF enzymes, with each monomer adopting an α/β fold and containing a zinc-binding site essential for catalysis. Comparative structural analysis reveals both conserved catalytic residues and species-specific variations in substrate-binding pockets that can be exploited for selective inhibitor design.
A methodological approach to characterizing these differences includes:
Homology modeling based on solved structures (such as B. subtilis IspF )
Multiple sequence alignment to identify conserved and variable regions
Expression and purification of recombinant V. vulnificus IspF for crystallographic studies
Structural comparison focusing on:
Active site architecture
Zinc coordination geometry
Conformational flexibility of the binding pocket
Surface properties affecting oligomerization
These structural differences may correlate with V. vulnificus's ability to adapt to changing environments during infection, potentially contributing to its pathogenicity .
For robust kinetic characterization of V. vulnificus IspF, a multi-method approach yields the most reliable data:
Continuous Spectrophotometric Assays:
Monitor CMP release via coupling with nucleotidase and phosphate detection
Track substrate consumption using coupled enzyme systems
Employ fluorescent substrate analogs for increased sensitivity
Discontinuous HPLC Analysis:
Separate and quantify reaction components at defined time points
Monitor both substrate depletion and product formation
Use isotopically labeled substrates for improved detection
Data Analysis Protocol:
Determine initial velocities across a range of substrate concentrations (0.1-10× Km)
Fit data to appropriate kinetic models (Michaelis-Menten, Hill, or substrate inhibition)
Analyze the effects of potential inhibitors using different inhibition models
Evaluate metal cofactor dependencies by varying zinc concentrations
Assess pH and temperature optima relevant to infection environments
This comprehensive approach enables correlation of enzymatic parameters with V. vulnificus pathogenicity, as the bacterium must adapt its metabolism during the transition from environmental reservoirs to human hosts .
The optimal expression system for V. vulnificus IspF requires balancing yield, solubility, and native conformation. Based on successful approaches with other bacterial enzymes and considering V. vulnificus's unique characteristics, the following methodological framework is recommended:
Expression System Selection:
Optimization Protocol:
Clone the ispF gene into vectors with various fusion tags (His6, MBP, SUMO)
Test expression under multiple conditions:
Induction temperature: 16°C, 25°C, 37°C
IPTG concentration: 0.1 mM, 0.5 mM, 1.0 mM
Media composition: LB, TB, auto-induction media
Duration: 4h, overnight, 24h
Evaluate protein solubility and activity for each condition
Scale up production using optimal parameters
This approach mirrors successful strategies used for studying other V. vulnificus proteins, where expression conditions significantly impacted protein quality and subsequent structural studies .
Purifying V. vulnificus IspF presents several challenges that require specific methodological solutions:
Solution: Include low concentrations of zinc (10-50 μM ZnCl2) in all buffers
Rationale: Zinc coordination stabilizes the trimeric assembly essential for activity
Solution: Optimize buffer conditions through thermal shift assays
Protocol:
Test various buffer compositions (pH 6.5-8.5)
Screen stabilizing additives (glycerol, trehalose, arginine)
Evaluate reducing agents (DTT, TCEP) at different concentrations
Determine optimal ionic strength (100-500 mM NaCl)
Solution: Multi-step purification strategy
IMAC (Ni-NTA) chromatography with imidazole gradient
Tag removal using precision protease
Anion exchange chromatography
Size exclusion chromatography to separate trimeric IspF
Solution: Activity-guided purification
Assay fractions at each purification step
Minimize exposure to potential inhibitors
Process samples rapidly at 4°C
Add substrate analogs to stabilize active conformation
This purification strategy addresses the specific challenges of V. vulnificus IspF while preserving the native structure necessary for accurate biochemical and structural characterization.
Successful crystallization of V. vulnificus IspF can be enhanced through targeted protein engineering approaches:
Surface Entropy Reduction:
Identify surface-exposed lysine and glutamate clusters using homology models
Replace selected clusters with alanines (e.g., K72A/K73A/K74A) to reduce conformational entropy
Screen engineered variants for improved crystallization propensity
Terminal Modifications:
Generate N- and C-terminal truncation series based on secondary structure predictions
Remove disordered regions that may impede crystal packing
Test each construct for solubility, activity, and crystallization behavior
Strategic Mutations:
Introduce mutations to disrupt crystal packing in undesirable orientations
Modify surface residues to promote specific crystal contacts
Replace cysteine residues to prevent non-native disulfide formation
Crystallization Facilitation Table:
| Engineering Approach | Methodology | Expected Outcome |
|---|---|---|
| Surface Entropy Reduction | Replace K/E clusters with alanines | Reduced surface entropy, improved crystal contacts |
| Truncation Constructs | Remove 5-10 residues from termini | Elimination of flexible regions |
| Binding Site Stabilization | Co-crystallize with substrates or inhibitors | Reduced conformational heterogeneity |
| Fusion Partners | Add T4 lysozyme or MBP | Additional crystal contacts, enhanced solubility |
These approaches have proven successful for crystallizing other bacterial enzymes, including IspF from B. subtilis, which yielded high-resolution structures suitable for detailed mechanistic analysis .
The active site of V. vulnificus IspF contains several critical residues that coordinate substrate binding and catalysis through a precise spatial arrangement:
Metal Coordination Site:
A zinc ion is coordinated by two histidines (typically His10 and His44, using B. subtilis numbering) and an aspartate (Asp11)
This zinc coordination is essential for positioning the substrate's phosphate groups and facilitating nucleophilic attack
Substrate Binding Residues:
Phosphate Binding:
Conserved lysine residues form salt bridges with substrate phosphates
Threonine and serine residues provide hydrogen bonding
Ribose Recognition:
Aspartate residues form hydrogen bonds with ribose hydroxyl groups
Hydrophobic residues create a pocket accommodating the ribose moiety
Cytidine Binding:
Aromatic residues (typically phenylalanine) provide π-stacking interactions with the cytosine ring
Backbone amides form hydrogen bonds with cytosine
Catalytic Mechanism Analysis:
The zinc ion coordinates the phosphate oxygen of the substrate
Conserved glutamate acts as a catalytic base, abstracting a proton
Nucleophilic attack leads to formation of the cyclic product
CMP is released as a leaving group
These structural features likely contribute to V. vulnificus's ability to adapt to changing host environments during infection, as precise metabolic regulation is essential for pathogen survival under stress conditions .
V. vulnificus IspF undergoes significant conformational changes during catalysis that are likely modulated by infection-specific conditions. Understanding these dynamics is crucial for both basic enzymology and drug development:
Key Conformational Transitions:
Open-to-closed transition upon substrate binding
Reorientation of mobile loops surrounding the active site
Subunit reorganization affecting intersubunit communication
Allosteric modulation by cellular metabolites
Methodological Approach to Study Conformational Changes:
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to detect regions of altered flexibility
Single-molecule FRET to monitor domain movements during catalysis
Molecular dynamics simulations to predict conformational trajectories
Crystallization in different states (apo, substrate-bound, product-bound)
Infection-Relevant Conformational Factors:
pH-dependent structural changes corresponding to different infection sites
Metal availability effects on active site geometry
Redox-sensitive conformational switches
Temperature-responsive structural elements
During infection, V. vulnificus experiences dramatic environmental changes, from estuarine environments to the human gastrointestinal tract and bloodstream . These transitions likely require adjustments in enzyme activity through conformational adaptation. For example, blood-specific conditions (temperature, pH, iron limitation) may trigger specific conformational states optimized for pathogen survival in this environment .
A comprehensive computational pipeline for identifying inhibitor binding sites in V. vulnificus IspF involves multiple complementary approaches:
Primary Site Identification:
Structure-based methods:
SiteMap analysis to identify binding pockets based on physical properties
FTMap to detect hot spots through fragment probe mapping
CASTp for geometric analysis of surface cavities
Sequence-based conservation analysis:
Prioritize sites showing differential conservation between bacterial and human enzymes
Identify species-specific pockets unique to V. vulnificus
Druggability Assessment:
Calculate physicochemical properties (volume, hydrophobicity, exposure)
Estimate binding energetics through molecular dynamics
Evaluate water displacement patterns using WaterMap
Score sites using machine learning algorithms trained on known druggable pockets
Dynamic Pocket Analysis:
Molecular dynamics simulations to identify transient pockets
Cryptic site discovery using enhanced sampling techniques
Allosteric network identification through community network analysis
Ensemble docking against multiple receptor conformations
Application to V. vulnificus IspF:
| Site Type | Computational Method | Validation Approach |
|---|---|---|
| Active site | Substrate-based pharmacophore modeling | Competitive inhibition assays |
| Allosteric sites | Normal mode analysis, MD simulations | Non-competitive inhibition profiles |
| Interface pockets | Protein-protein interface analysis | Oligomerization disruption assays |
| Species-specific cavities | Comparative modeling with other bacterial IspFs | Selectivity testing |
This computational pipeline provides multiple targeting opportunities for developing selective inhibitors against V. vulnificus IspF, potentially addressing the need for new approaches against this pathogen with high mortality rates .
The relationship between IspF activity and virulence factor expression in V. vulnificus represents a complex regulatory network that can be systematically investigated:
Methodological Approach:
Generate a conditional ispF mutant with titratable expression
Quantify virulence factor production at different IspF activity levels:
Perform transcriptomic analysis to identify co-regulated genes
Use chromatin immunoprecipitation to identify direct regulatory interactions
Potential Regulatory Mechanisms:
Metabolic coupling through shared precursors
Isoprenoid-dependent signaling pathways
Influence on membrane properties affecting secretion systems
Integration with the ToxRS system, known to regulate virulence gene expression
Research suggests that V. vulnificus virulence factors are tightly regulated and produced at specific times during infection . IspF activity likely influences this regulation through metabolic integration with pathways controlling virulence gene expression. The isoprenoid products of the MEP pathway contribute to membrane structure, which in turn affects the function of membrane-associated virulence determinants like the ToxRS regulatory system .
Developing selective inhibitors against V. vulnificus IspF requires a systematic approach addressing multiple aspects of drug design:
Target Validation:
Confirm essentiality of IspF for V. vulnificus growth and virulence
Determine minimum inhibition level required for antimicrobial effect
Assess potential for resistance development through mutation rate studies
Structure-Based Design Strategy:
Obtain high-resolution structures of V. vulnificus IspF (similar to B. subtilis IspF )
Perform comparative analysis with human isoprenoid-synthesizing enzymes
Identify unique structural features of V. vulnificus IspF for selective targeting
Design initial compounds targeting:
The active site zinc-binding region
Species-specific substrate binding pockets
Allosteric sites affecting trimer formation
Compound Optimization Pipeline:
| Stage | Methodology | Criteria |
|---|---|---|
| Initial screening | Fragment-based screening, virtual screening | Binding affinity, novelty |
| Hit expansion | Medicinal chemistry, structure-activity relationships | Improved potency, selectivity |
| Lead optimization | Pharmacokinetic studies, toxicity assessment | Reduced off-target effects |
| Candidate selection | In vivo efficacy in infection models | Therapeutic index, dosing |
Delivery Considerations:
Design compounds that can penetrate V. vulnificus's Gram-negative cell envelope
Consider prodrug approaches to enhance bioavailability
Evaluate combination strategies with membrane permeabilizers
This approach addresses the urgent need for new therapeutics against V. vulnificus, which causes rapidly progressing infections with mortality rates exceeding 50% .
V. vulnificus must adapt to multiple host defense mechanisms during infection, and IspF function likely plays a critical role in this adaptation:
Immune Pressure Adaptation Mechanisms:
Membrane Remodeling:
IspF-derived isoprenoids contribute to membrane fluidity and integrity
Altered membrane composition affects resistance to antimicrobial peptides
Capsular polysaccharide (CPS) integration requires isoprenoid-linked carriers
Oxidative Stress Response:
Isoprenoid-derived antioxidants protect against reactive oxygen species
MEP pathway modulation may compensate for oxidative damage
Carotenoid synthesis (downstream of MEP pathway) provides additional protection
Immune Evasion:
Methodological Investigation Approach:
Challenge ispF conditional mutants with specific immune components:
Neutrophils and neutrophil extracellular traps (NETs)
Serum complement
Antimicrobial peptides
Macrophages under different activation states
Monitor transcriptional responses of ispF and the MEP pathway during immune challenge
Correlate IspF activity with known immune evasion mechanisms:
This research would elucidate how metabolic adaptations mediated by IspF contribute to V. vulnificus's remarkable virulence and ability to cause fatal septicemia in susceptible hosts .
CRISPR-Cas9 technology enables sophisticated genetic manipulations to dissect IspF function in V. vulnificus:
Genome Editing Applications:
Domain-specific mutations:
Create precise amino acid substitutions in catalytic residues
Modify zinc-binding motifs to alter metal coordination
Engineer interface residues to disrupt trimerization
Regulatory element analysis:
Edit promoter regions to identify regulatory elements
Modify untranslated regions affecting mRNA stability
Create reporter fusions to monitor expression dynamics
CRISPR Interference (CRISPRi) Applications:
Temporal control of ispF expression:
Titrate expression levels using inducible dCas9 systems
Study metabolic consequences of partial IspF inhibition
Identify minimum expression threshold for viability
Spatial regulation studies:
Use tissue-specific promoters to drive dCas9 expression in animal models
Determine organ-specific requirements for IspF activity
CRISPR Activation (CRISPRa) Applications:
Overexpression studies:
Upregulate ispF to determine effects on isoprenoid flux
Investigate potential toxicity of MEP pathway intermediate accumulation
Study compensatory responses to IspF overexpression
Multiplexed CRISPR Applications:
Simultaneous targeting of multiple MEP pathway enzymes
Creation of synthetic genetic interaction maps
Identification of genetic suppressors of ispF deficiency
This CRISPR-based approach provides unprecedented precision for studying IspF function in the context of V. vulnificus metabolism and pathogenesis, potentially revealing new aspects of this enzyme's role in bacterial adaptation during infection .
A comprehensive structural analysis of V. vulnificus IspF would provide valuable insights for both basic understanding and drug development:
Structural Determination Methodology:
X-ray crystallography approach:
Cryo-EM analysis:
Prepare vitrified samples of purified IspF
Collect high-resolution image data
Perform 3D reconstruction to generate electron density maps
Build and refine atomic models
Comparative Structural Analysis:
Structure-Function Correlations:
Identify conformational changes associated with substrate binding
Map conserved catalytic residues and species-specific variations
Analyze oligomerization interfaces and potential for disruption
Predict binding sites for rational inhibitor design
This structural knowledge would facilitate understanding of IspF's role in V. vulnificus metabolism during infection and provide a foundation for structure-based drug design targeting this essential enzyme .
V. vulnificus transitions through dramatically different environments during infection, potentially requiring post-translational modification (PTM) of key enzymes like IspF to adapt:
Potential Post-Translational Modifications:
Phosphorylation:
Regulates enzyme activity in response to environmental signals
May be controlled by two-component systems sensing host conditions
Can affect subunit interactions and catalytic efficiency
Oxidative Modifications:
Cysteine oxidation under oxidative stress conditions
Potential redox sensing mechanism during host immune response
May protect key residues during oxidative burst by phagocytes
Metal Ion Coordination Changes:
Zinc availability fluctuates during infection (competition with host)
Alternative metal incorporation may affect catalytic properties
Host nutritional immunity strategies target metal-dependent enzymes
Methodological Investigation Approach:
| PTM Type | Detection Method | Functional Analysis |
|---|---|---|
| Phosphorylation | Phosphoproteomics, Phos-tag gels | Site-directed mutagenesis to Ala or Asp |
| Oxidation | Redox proteomics, Mass spectrometry | Differential alkylation, redox titration |
| Metal exchange | ICP-MS, X-ray absorption spectroscopy | Activity assays with different metals |
| Acetylation | Acetylome analysis | Mutation to mimic acetylated/deacetylated states |
Environmental Correlation:
Compare PTM profiles between:
Environmental growth conditions (estuarine-like)
Gastrointestinal simulation
Serum-exposed bacteria
Intracellular bacteria within macrophages
Correlate PTM patterns with:
Virulence gene expression
Metabolic flux through the MEP pathway
Resistance to host defense mechanisms
This research would provide insights into how V. vulnificus adapts its central metabolism during the infection process, potentially revealing new vulnerabilities that could be exploited therapeutically .