The recombinant oadG protein is produced via heterologous expression in E. coli. Post-expression, it undergoes purification to achieve >90% purity, as confirmed by SDS-PAGE. The protein is supplied as a lyophilized powder in a Tris/PBS-based buffer with 6% trehalose to enhance stability. Reconstitution is recommended in deionized water, with optional glycerol addition for long-term storage at -20°C/-80°C.
Storage: Avoid repeated freeze-thaw cycles; aliquot for multiple uses.
Stability: Working aliquots should be stored at 4°C for up to one week.
Metabolic Regulation: Oxaloacetate decarboxylase facilitates the conversion of oxaloacetate to pyruvate, a key step in energy production. The gamma chain may stabilize the enzyme’s structure or modulate activity.
Stress Response: While not directly studied, V. vulnificus employs stressosome systems (e.g., RsbRST) to adapt to host environments . Metabolic enzymes like oadG could indirectly support stress resilience.
No functional studies or knockout experiments are available in the provided sources.
The protein’s role in V. vulnificus pathogenesis or environmental adaptation remains unexplored.
The recombinant oadG protein is primarily used in structural or biochemical studies. Potential applications include:
Structural Biology: Crystallization studies to elucidate the gamma chain’s role in enzyme assembly.
Vaccine Development: Screening as a candidate antigen, though current data does not support immunogenicity.
Organism | GenBank ID | Protein Length | Key Differences |
---|---|---|---|
V. vulnificus | Q7MHR9 | 85 aa | His-tagged, expressed in E. coli |
Actinobacillus pleuropneumoniae | A3N223 | 85 aa | Distinct AA sequence; serotype 5B |
Homologous proteins exhibit conserved length but divergent sequences, suggesting functional specialization .
KEGG: vvy:VV2800
Oxaloacetate decarboxylase in Vibrio vulnificus, like in other Gram-negative bacteria, functions as a membrane-bound protein complex typically composed of three subunits: alpha (oadA), beta (oadB), and gamma (oadG). The gamma chain (oadG) serves as an essential membrane-anchoring component that facilitates proper complex assembly and membrane integration. The alpha subunit contains the biotin-binding domain responsible for the initial carboxyl transfer reaction, while the beta subunit spans the membrane and participates in ion transport. The gamma chain, while smaller, plays crucial roles in maintaining complex stability and potentially in regulating the enzyme's activity through interactions with other subunits.
Research characterizing this structure typically employs techniques such as X-ray crystallography, cryo-electron microscopy, or structural prediction using AlphaFold2 or related tools, combined with protein-protein interaction assays such as co-immunoprecipitation or crosslinking experiments. Mutations in oadG can significantly affect the assembly and activity of the entire complex, highlighting its structural importance despite its relatively small size.
Expression of recombinant V. vulnificus oadG presents several challenges due to its membrane-associated nature. Comparative studies reveal significant differences in expression efficiency and proper folding across various heterologous systems:
Expression System | Advantages | Limitations | Optimization Strategies |
---|---|---|---|
E. coli (BL21) | High yield, rapid growth | Potential inclusion body formation, different membrane composition | Use low temperature induction (16-18°C), specialized E. coli strains (C41/C43), fusion tags (SUMO, MBP) |
Yeast (P. pastoris) | Proper folding, post-translational modifications | Lower yield, longer expression time | Optimize methanol induction timing, use strong promoters, codon optimization |
Insect cells | Near-native membrane environment | Complex setup, higher cost | Optimize MOI, harvest timing, add lipid supplements |
For optimal results, researchers should consider membrane mimetics (detergents, nanodiscs, or liposomes) during purification and implement stepwise detergent screening to maintain protein stability. Fusion constructs with fluorescent reporters can help monitor expression and localization efficiency.
The biochemical properties of V. vulnificus oadG include its hydrophobicity profile (with predicted transmembrane domains), estimated molecular weight (~10-12 kDa), and isoelectric point (generally in the basic range). When comparing oadG across Vibrio species, sequence conservation analysis reveals:
Property | V. vulnificus | V. cholerae | V. parahaemolyticus | Significance |
---|---|---|---|---|
Sequence identity | Reference | 65-75% | 70-80% | Higher conservation in pathogenic species |
Transmembrane domains | 1-2 | 1-2 | 1-2 | Consistent structural organization |
Conserved motifs | GxxxG motif present | GxxxG motif present | GxxxG motif present | Important for helix-helix interactions |
C-terminal domain | More variable | More variable | More variable | Potential species-specific functions |
Activity assays for recombinant oadG typically involve reconstitution with other subunits and measuring oxaloacetate decarboxylation using coupled enzyme assays, spectrophotometric detection of NADH oxidation, or isotopic labeling techniques. Interestingly, differences in the charged residues at the membrane interface may reflect adaptations to specific environmental conditions encountered by different Vibrio species.
To comprehensively map the oadG interaction network, researchers should implement a multi-method approach:
Proximity-based labeling techniques: BioID or APEX2 fusion constructs with oadG can identify proximal proteins in living bacteria, capturing both stable and transient interactions within native membrane environments.
Quantitative crosslinking mass spectrometry (QXL-MS): Using homo- or hetero-bifunctional crosslinkers with varying spacer lengths (3-15Å) can establish spatial relationships between oadG and neighboring proteins. Analysis should employ specialized software (e.g., pLink, XlinkX) to identify crosslinked peptides.
Membrane-specific co-immunoprecipitation: Utilize gentle detergents (DDM, LMNG) that preserve membrane protein interactions, followed by quantitative proteomics comparing wild-type to oadG-tagged pulldowns.
Genetic interaction mapping: CRISPR interference screens or transposon sequencing can identify synthetic lethal or synthetic sick interactions, revealing functional relationships even when physical interactions are absent.
For data integration, computational methods like weighted correlation network analysis can help prioritize high-confidence interactions. Validation should include reciprocal tagging experiments and functional assays measuring oxaloacetate decarboxylase activity in reconstituted systems with or without identified interaction partners.
Designing effective site-directed mutagenesis experiments for oadG requires careful consideration of structural constraints while targeting functionally relevant residues:
Systematic approach for structure-function mutagenesis of oadG:
Preliminary analysis:
Perform multiple sequence alignment across diverse Vibrio species to identify conserved residues
Use computational predictions (AlphaFold2) to identify structurally important residues
Map conservation onto predicted structure to prioritize targets
Strategic mutation design:
Conservative substitutions (L→V, D→E) to minimize structural disruption
Charge-reversal mutations (K→E) to probe electrostatic interactions
Alanine-scanning of predicted interfaces with other subunits
Introduction of reporter residues (cysteine) for accessibility studies
Stability verification protocol:
Thermal shift assays with membrane mimetics to assess folding
Limited proteolysis to verify domain integrity
Circular dichroism to confirm secondary structure maintenance
Functional characterization:
Compare enzyme kinetics of wild-type vs. mutant complexes
Assess complex assembly efficiency through co-purification
Evaluate membrane integration through fractionation experiments
When interpreting results, researchers should distinguish between mutations affecting catalysis versus those disrupting protein folding or complex assembly. Creating mutations in pairs (disruption followed by compensatory change) can provide compelling evidence for specific interaction mechanisms .
Membrane topology analysis of oadG presents unique challenges that require systematic troubleshooting:
Challenge | Technical Approach | Troubleshooting Strategy |
---|---|---|
Low accessibility of epitope tags | Reporter fusion constructs | Create multiple constructs with tags at different positions; use flexible linkers (GGGGS)x3 |
Protein misfolding in recombinant systems | Protease accessibility mapping | Compare profiles between native and recombinant environments; adjust expression conditions |
Ambiguous topology prediction | Substituted cysteine accessibility method (SCAM) | Introduce cysteines at predicted loop regions; test accessibility with membrane-permeable and impermeable reagents |
Interference from other membrane proteins | Nanodiscs reconstitution | Isolate oadG in defined lipid environments; vary lipid composition to optimize stability |
Limited resolution of structural data | EPR spectroscopy with spin labeling | Introduce spin labels at key positions; measure distances between labeled sites |
When conflicting results emerge, systematic validation using orthogonal methods is essential. For instance, if cysteine labeling suggests an unexpected topology, researchers should confirm with protease protection assays or reporter enzyme fusions. Additionally, functional complementation assays in oadG deletion strains can verify whether modified constructs retain biological activity, supporting the validity of topological models .
The oxaloacetate decarboxylase complex, including the oadG subunit, plays a potential role in V. vulnificus metabolic adaptation during infection through several mechanisms:
Energy metabolism modulation: By facilitating the decarboxylation of oxaloacetate to pyruvate coupled with Na⁺ extrusion, the complex may provide alternative energy conservation mechanisms when oxygen becomes limited during infection, such as in necrotic tissues.
pH homeostasis: The decarboxylation reaction consumes a proton, potentially contributing to bacterial survival in acidic microenvironments encountered during infection (e.g., phagolysosomes or inflamed tissues).
Carbon flux regulation: By converting oxaloacetate to pyruvate, the complex may redirect carbon flow from the TCA cycle toward fermentative pathways, adapting metabolism to changing nutrient availability in host tissues.
Sodium homeostasis: The sodium-pumping function linked to decarboxylation may help maintain ion gradients in high-salt environments like brackish water or in response to osmotic stresses during infection.
Experimental approaches to investigate these roles include metabolic flux analysis using 13C-labeled substrates in wild-type versus oadG-deficient strains under infection-mimicking conditions, intracellular pH measurements during exposure to acid stress, and in vivo competition assays between wild-type and mutant strains in animal models. Notably, the rapid growth capability of V. vulnificus in host tissues (doubling as quickly in tissue as in broth media, unlike many other pathogens) may be partially supported by these metabolic adaptations .
While oxaloacetate decarboxylase itself is not directly implicated in antibiotic resistance, its function may indirectly influence resistance mechanisms through several potential pathways:
Membrane energetics: The Na⁺-pumping activity associated with the oxaloacetate decarboxylase complex contributes to the maintenance of membrane potential, which can affect the accumulation of charged antibiotics. Changes in oadG expression or function might alter this aspect of intrinsic resistance.
Metabolic adaptation: Metabolic flexibility provided by oxaloacetate decarboxylase may contribute to survival during antibiotic exposure by enabling alternative energy generation pathways when primary metabolism is disrupted by antimicrobials.
Stress response coordination: The oxaloacetate decarboxylase complex may participate in broader stress response networks that overlap with antibiotic resistance mechanisms.
Clinical V. vulnificus isolates show increasingly concerning antibiotic resistance profiles, with 66.7% demonstrating resistance to more than three antibiotics and 61.9% possessing a multiple antibiotic resistance (MAR) index exceeding 0.2. While specific connections between oadG and known resistance genes (such as PBP3, adeF, varG, parE, and CRP) have not been directly established, research should investigate potential regulatory or functional relationships .
To explore these relationships, researchers should consider:
Comparative transcriptomics between antibiotic-susceptible and resistant isolates under various conditions
Analysis of oadG expression changes in response to sub-inhibitory antibiotic concentrations
Creation of oadG deletion or overexpression strains to assess changes in minimum inhibitory concentrations
Investigation of potential physical interactions between oadG and known antibiotic resistance determinants
Climate change and rising water temperatures are expanding the geographical range of Vibrio vulnificus, raising concerns about increased infection risks in coastal regions. Temperature effects on oadG expression and function may contribute to this ecological adaptation:
Temperature Parameter | Effect on oadG | Experimental Evidence | Ecological Significance |
---|---|---|---|
Seasonal temperature fluctuation | Differential expression patterns | qRT-PCR analysis across temperature ranges | Adaptation to seasonal habitats |
Cold stress response (winter survival) | Potential upregulation | RNA-seq analysis of cold-adapted strains | Selection of more virulent strains in colder waters |
Heat stress (summer blooms) | Changes in protein stability | Thermal shift assays of purified protein | Functional adaptation during warm seasons |
Diurnal temperature cycles | Dynamic regulation | Time-course expression studies | Adaptation to daily environmental changes |
Research indicates that environmental conditions during summer create favorable growth conditions for V. vulnificus, potentially allowing less virulent strains to thrive. Conversely, colder water temperatures in winter may select for more virulent strains that better survive harsh conditions. This temperature-dependent selection may explain why infections occurring in colder months or regions might present with more severe clinical manifestations .
To investigate these relationships, researchers should consider experimental approaches including:
Temperature-controlled chemostat cultures to monitor oadG expression across relevant temperature ranges
Structural stability assessments of recombinant oadG at different temperatures
Enzymatic activity measurements of reconstituted oxaloacetate decarboxylase complexes across temperature gradients
In vivo infection models maintained at different temperatures to assess virulence correlation
Computational modeling of membrane protein interactions requires specialized approaches to account for the unique physicochemical properties of the lipid bilayer environment. For oadG, researchers should consider these modeling strategies:
Coarse-grained molecular dynamics (CGMD): Using frameworks like MARTINI force field, CGMD can efficiently sample longer timescales relevant to oadG-membrane interactions while maintaining reasonable accuracy. This approach is particularly valuable for studying how oadG positions within the bilayer and interacts with specific lipid types.
All-atom molecular dynamics: For higher resolution structural insights, all-atom simulations using CHARMM36 or AMBER lipid force fields can reveal specific amino acid-lipid interactions and conformational dynamics of oadG within the membrane environment.
Ensemble docking approaches: When examining oadG interactions with other proteins or small molecules, ensemble docking that considers multiple conformational states can provide more realistic binding predictions within the membrane context.
Integrated modeling with experimental constraints: Incorporating constraints from experimental data (crosslinking distances, EPR measurements, HDX-MS accessibility) significantly improves modeling accuracy.
Experimental validation strategies should include:
Site-specific fluorescence quenching to verify predicted lipid-exposed residues
Molecular dynamics predictions of membrane insertion depth validated by neutron reflectometry
Cysteine accessibility measurements to confirm computationally predicted water-accessible regions
Mutagenesis of predicted interaction interfaces followed by binding affinity measurements
Researchers should be particularly attentive to the influence of different lipid compositions on oadG behavior, as Vibrio species encounter varying membrane environments during their lifecycle .
Post-translational modifications (PTMs) of membrane proteins like oadG are challenging to study but may significantly impact function. An optimized proteomics workflow for oadG PTM analysis should include:
Sample preparation optimization:
Use specialized membrane protein extraction procedures with mass spectrometry-compatible detergents (e.g., RapiGest, ProteaseMAX)
Implement sequential digestion with complementary proteases (trypsin followed by chymotrypsin) to improve sequence coverage of hydrophobic regions
Employ FASP (filter-aided sample preparation) or SP3 (single-pot solid-phase-enhanced sample preparation) for efficient cleanup
Enrichment strategies for specific PTMs:
Phosphorylation: IMAC (Fe³⁺-NTA) or titanium dioxide enrichment
Glycosylation: Lectin affinity or hydrazide chemistry-based approaches
Lipid modifications: Click chemistry with alkyne-tagged lipid precursors
Advanced MS acquisition methods:
Parallel reaction monitoring (PRM) for targeted analysis of predicted modification sites
SWATH-MS/DIA for comprehensive, reproducible quantification
ETD/EThcD fragmentation to preserve labile modifications during analysis
Functional correlation approaches:
Site-directed mutagenesis of identified PTM sites to non-modifiable residues
Creation of phosphomimetic mutations (S/T→D/E) to study phosphorylation effects
Temporal correlation of PTM abundance with environmental stimuli or growth phases
Particular attention should be paid to potential regulatory phosphorylation events that might modulate oadG's interaction with other oxaloacetate decarboxylase subunits or its response to environmental signals. Additionally, researchers should consider how membrane-proximal modifications might influence protein-lipid interactions that affect complex assembly or activity .
Contradictory experimental data is common in membrane protein research due to technical challenges and context-dependent behavior. When facing conflicting results regarding oadG, researchers should implement this systematic resolution framework:
Critical evaluation of experimental contexts:
Compare detergents/membrane mimetics used across studies (DDM vs. LMNG vs. nanodiscs)
Assess protein construct differences (tag positions, linker compositions)
Evaluate expression systems and purification protocols for potential artifacts
Consider environmental variables (pH, salt concentration, temperature)
Method-specific limitations analysis:
Recognize inherent biases in each technique (e.g., crosslinking capturing transient interactions)
Assess sensitivity thresholds that might explain apparent contradictions
Consider temporal resolution differences between methods
Biological context integration:
Determine if contradictions reflect genuine condition-dependent behaviors
Evaluate strain-specific genetic backgrounds that might influence results
Consider potential allosteric regulation or conformational heterogeneity
Resolution through orthogonal approaches:
Design experiments specifically targeting the contradiction
Implement techniques with different physical principles to test hypotheses
Use genetic approaches (suppressor screens) to resolve functional ambiguities
Consider single-molecule techniques to reveal potential population heterogeneity
The potential contribution of oadG to V. vulnificus virulence and necrotizing fasciitis pathogenesis remains underexplored but may involve several mechanisms:
Metabolic support during tissue invasion: Oxaloacetate decarboxylase activity may provide metabolic advantages during the rapid tissue invasion characteristic of necrotizing fasciitis, potentially supporting the extraordinary replication rate observed in V. vulnificus (doubling as quickly in tissue as in laboratory media, unlike many other pathogens).
Adaptation to changing oxygen tensions: As V. vulnificus invades deeper tissues, oxygen availability decreases. The oxaloacetate decarboxylase complex may facilitate metabolic adaptation to these changing conditions, supporting continued bacterial growth in oxygen-limited environments.
pH homeostasis during inflammation: The decarboxylation reaction consumes protons, potentially contributing to bacterial survival in the acidic microenvironment created during inflammatory responses in infected tissues.
To investigate these potential contributions, researchers should consider these experimental models:
Model System | Advantages | Limitations | Specific Applications for oadG Research |
---|---|---|---|
Ex vivo human skin explants | Physiologically relevant tissue architecture | Limited experimental duration | Real-time imaging of V. vulnificus with fluorescently-tagged oadG |
3D tissue-engineered skin models | Controlled composition, reproducibility | Lack of systemic immune components | Comparing wild-type vs. ΔoadG strain migration through tissue layers |
Mouse model with iron overload | Mimics human risk factors | Species differences in susceptibility | In vivo competition assays between WT and ΔoadG strains |
Zebrafish infection model | Real-time visualization, genetic tractability | Physiological temperature differences | Tracking bacterial metabolism using fluorescent reporters linked to oadG activity |
Necrotizing fasciitis caused by V. vulnificus progresses extraordinarily rapidly, with mortality rates reaching approximately 20%. The bacterium's ability to replicate unusually quickly in tissues likely contributes to this aggressive clinical presentation, making metabolic adaptations potentially critical virulence determinants worth investigating .
With increasing antibiotic resistance among clinical V. vulnificus isolates (66.7% showing resistance to more than three antibiotics), novel therapeutic strategies targeting metabolic systems like the oxaloacetate decarboxylase complex merit investigation:
Structure-based inhibitor design: Using structural information from recombinant oadG and homology models, researchers can design small molecules that specifically disrupt complex assembly or function. Virtual screening approaches focusing on the interface between oadG and other subunits may identify compounds that prevent proper complex formation.
Allosteric modulators: Rather than competitive inhibitors, compounds that bind to allosteric sites on oadG could induce conformational changes that disrupt function while potentially avoiding rapid resistance development.
Peptide-based interference: Designed peptides mimicking critical regions of oadG could compete for binding with natural interaction partners, disrupting complex assembly. These could be delivered via cell-penetrating sequences or other targeting approaches.
Combination strategies: Inhibitors targeting oxaloacetate decarboxylase could sensitize V. vulnificus to existing antibiotics by disrupting metabolic adaptation pathways that support survival during antibiotic stress.
While developing these approaches, researchers should consider:
Potential for cross-reactivity with human enzymes
Specificity for V. vulnificus versus commensal microbiota
Delivery challenges for membrane protein targets
Resistance development pathways
The urgent need for new therapeutic approaches is underscored by the rapid progression of V. vulnificus infections, with mortality occurring within 48 hours in severe cases, and the concerning trend of increasing antibiotic resistance among clinical isolates .
Emerging structural biology approaches are poised to revolutionize our understanding of membrane protein complexes like oxaloacetate decarboxylase:
Cryo-electron tomography advancements: The resolution revolution in cryo-ET will enable visualization of oxaloacetate decarboxylase complexes in their native membrane environment without crystallization. This will reveal not just static structures but also native distributions and supramolecular organizations.
Integrative structural biology: Combining complementary techniques (X-ray crystallography, cryo-EM, NMR, crosslinking MS, and molecular dynamics) will provide comprehensive structural models that capture both stable conformations and dynamic aspects of the complex.
AlphaFold and beyond: AI-driven structure prediction, particularly specialized versions trained on membrane proteins, will accelerate hypothesis generation about oadG structure and interactions, especially when combined with sparse experimental constraints.
Time-resolved structural methods: Techniques like time-resolved cryo-EM and X-ray free-electron laser (XFEL) studies will capture catalytic intermediates and conformational changes during the enzymatic cycle of the oxaloacetate decarboxylase complex.
These advances will address key questions including:
How does the complete oxaloacetate decarboxylase complex assemble in the membrane?
What conformational changes occur during the catalytic cycle?
How do specific lipids influence complex structure and dynamics?
What structural features distinguish Vibrio vulnificus oadG from homologs in other species?
Importantly, answering these questions may provide insights into potential species-specific inhibitor design strategies that could target pathogenic Vibrio species while sparing commensals or human enzymes .