Recombinant Vibrio vulnificus 30S ribosomal protein S3 (rpsC) is a genetically engineered version of the protein S3, which is a component of the 30S ribosomal subunit in bacteria. This protein plays a crucial role in protein synthesis by facilitating the translation of messenger RNA (mRNA) into proteins. The recombinant form of this protein is produced using biotechnological methods, typically in host organisms like Escherichia coli, yeast, or mammalian cells .
The rpsC gene encodes the 30S ribosomal protein S3, which is essential for the structural integrity and function of the ribosome. Beyond its role in translation, ribosomal proteins can have extraribosomal functions, such as involvement in DNA repair and regulation of cell death pathways, although these functions are more commonly associated with eukaryotic ribosomal proteins .
Recombinant Vibrio vulnificus 30S ribosomal protein S3 (rpsC) is produced with high purity, typically greater than or equal to 85%, as determined by SDS-PAGE (Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis) . This level of purity is crucial for research applications, ensuring that the protein's functions and interactions can be studied accurately without contamination from other proteins.
This recombinant protein can be used in various research applications, including:
Structural Studies: Understanding the structure of the 30S ribosomal subunit and how it interacts with other components.
Antibiotic Resistance: Studying mutations in ribosomal proteins that confer resistance to antibiotics like tigecycline, which targets the 30S subunit .
Protein Synthesis: Investigating the role of S3 in the translation process and its potential extraribosomal functions.
Characteristic | Description |
---|---|
Gene Name | rpsC |
Protein Name | 30S Ribosomal Protein S3 |
Host Organisms | E. coli, Yeast, Baculovirus, Mammalian Cells |
Purity | ≥ 85% (SDS-PAGE) |
Applications | Structural studies, antibiotic resistance research, protein synthesis studies |
While specific research findings on Recombinant Vibrio vulnificus 30S ribosomal protein S3 (rpsC) might be limited, studies on similar proteins highlight their importance in ribosome function and potential roles in antibiotic resistance. For example, mutations in ribosomal proteins can alter the binding affinity of antibiotics to the ribosome, affecting their efficacy .
KEGG: vvy:VV0381
The 30S ribosomal protein S3 (rpsC) in Vibrio vulnificus is a component of the small ribosomal subunit essential for protein translation. In bacterial systems, rpsC participates in mRNA binding and helps maintain the structural integrity of the ribosome. Beyond its canonical role in translation, recent research suggests rpsC may function as an extraribosomal protein involved in stress responses, similar to how RpoS functions as a global regulator that helps V. vulnificus acquire resistance against various stresses and express virulence factors .
While direct evidence linking rpsC to pathogenicity is still emerging, research on other bacterial pathogens suggests that ribosomal proteins can play extraribosomal roles in virulence. In V. vulnificus specifically, proteins involved in fundamental cellular processes often demonstrate dual functionality in pathogenesis. Similar to how RpoS regulates virulence factors such as elastase and exoproteases in V. vulnificus , rpsC may contribute to pathogenicity through regulation of stress responses or interaction with host factors during infection. V. vulnificus causes deadly septicemia with rapid pathogenic progression and high mortality rates , making all potential virulence factors important research targets.
The rpsC gene in V. vulnificus is part of the core genome conserved across strains. Similar to other essential genes in this pathogen, rpsC likely features promoter elements that respond to environmental cues. The genomic context of rpsC may involve operonic organization with other ribosomal protein genes, a common arrangement in bacteria. Genomic analyses have revealed high recombination rates and frequent exchange of mobile genetic elements between V. vulnificus populations , suggesting that rpsC sequence variation should be examined across clinical versus environmental isolates to identify potential clinical-associated alleles (CAAs).
For recombinant V. vulnificus rpsC expression, E. coli-based systems typically yield the best results. The pET expression system with BL21(DE3) or Rosetta strains has proven particularly effective due to the codon optimization requirements for Vibrio genes. Expression should be induced with 0.5-1.0 mM IPTG at lower temperatures (16-25°C) to enhance protein solubility. Adding solubility tags such as MBP (maltose-binding protein) or SUMO can further improve yield and solubility. This approach mirrors established protocols for expressing other V. vulnificus proteins, where careful control of expression conditions has been shown to significantly impact protein functionality and yield.
Purification of recombinant V. vulnificus rpsC presents several challenges, including:
Tendency to co-purify with host ribosomal components and RNA
Formation of inclusion bodies when overexpressed
Potential instability in standard buffer conditions
A recommended purification protocol involves:
Initial capture using affinity chromatography (His-tag or tag-specific column)
RNase treatment to remove bound nucleic acids
Ion exchange chromatography to separate charged variants
Size exclusion chromatography for final polishing
Buffer optimization is critical, with best results typically achieved using 50 mM Tris-HCl (pH 7.5), 300 mM NaCl, 5% glycerol, and 1 mM DTT. Including low concentrations of detergents (0.05% Tween-20) may help prevent protein aggregation.
Optimizing solubility for recombinant V. vulnificus rpsC requires a multi-faceted approach:
Expression temperature reduction to 16-18°C
Co-expression with molecular chaperones (GroEL/GroES system)
Addition of osmolytes like glycerol (5-10%) or sucrose (2-5%) to expression media
Use of lysis buffers containing mild solubilizing agents:
1-2% sarkosyl followed by dilution
0.5-1 M urea (non-denaturing concentration)
0.1-0.5% Triton X-100
For particularly challenging preparations, a denaturation-refolding approach may be necessary, using 6 M guanidine-HCl for initial solubilization followed by step-wise dialysis into native buffer conditions. This approach must be carefully optimized, as incorrect refolding can affect protein function.
Analyzing structure-function relationships of V. vulnificus rpsC requires combining structural approaches with functional assays:
Structural Methods:
Functional Assays:
In vitro translation assays using purified V. vulnificus ribosomes
RNA binding assays (electrophoretic mobility shift assays, filter binding)
Ribosome assembly studies
Stress response induction measurements
The combination of these approaches allows correlation between specific structural elements and functional activities, similar to approaches used to study other V. vulnificus proteins like RpoS, where binding capabilities were assessed using electrophoretic mobility shift assays (EMSA) .
V. vulnificus rpsC interacts with mRNA primarily through its N-terminal domain, which contains basic residues that engage with the negatively charged phosphate backbone of RNA. This interaction is critical for mRNA binding during translation initiation and elongation. Within the ribosome, rpsC forms multiple protein-protein interactions with neighboring ribosomal proteins, particularly with proteins S4, S5, and S10 in the platform of the 30S subunit.
These interactions can be studied using:
Cross-linking studies followed by mass spectrometry
Hydrogen-deuterium exchange mass spectrometry
Site-directed mutagenesis of putative interaction sites
Fluorescence resonance energy transfer (FRET) to measure distances between components
Comparing these interaction patterns with those seen in other bacterial species can provide insight into V. vulnificus-specific translation mechanisms and potential antibiotic targets.
Post-translational modifications (PTMs) in native V. vulnificus rpsC may include:
Methylation
Acetylation
Phosphorylation
Hydroxylation
These modifications likely play regulatory roles in ribosome assembly, translation efficiency, and potentially extraribosomal functions. Identifying these PTMs requires:
Mass spectrometry-based approaches:
Bottom-up proteomics with enrichment strategies for specific PTMs
Top-down proteomics for intact protein analysis
Middle-down approaches for large peptide analysis
Site-specific detection methods:
Western blotting with modification-specific antibodies
Chemical labeling of specific modifications
Comparative analysis between different growth conditions and stress responses
PTM patterns may vary between environmental and clinical isolates, potentially contributing to strain-specific virulence attributes, similar to how clinical-associated alleles (CAAs) have been identified in other V. vulnificus proteins .
The expression of rpsC in V. vulnificus likely undergoes dynamic regulation during infection. Based on patterns observed with other V. vulnificus proteins:
Initial infection stage: rpsC expression may remain consistent with normal translation requirements
Host adaptation phase: Potential upregulation to support increased protein synthesis needs during rapid proliferation
Stress response phase: Possible differential regulation when encountering host immune defenses, similar to how RpoS levels increase when cells enter stationary phase or encounter stress conditions
Monitoring these expression changes requires:
qRT-PCR with infection time-course samples
Proteomics analysis of V. vulnificus during infection
Reporter gene constructs (such as rpsC::luxAB) to track expression in real-time models
Western blot analysis with rpsC-specific antibodies
Research investigating RpoS has shown that expression patterns can be significantly affected by environmental conditions and stress factors , and similar principles likely apply to rpsC expression.
While direct evidence for extraribosomal roles of rpsC in V. vulnificus virulence is still emerging, several experimental approaches can assess this potential:
Protein-protein interaction studies identifying non-ribosomal binding partners
Conditional knockdown studies examining virulence phenotypes independent of translation effects
Localization studies showing non-ribosomal distribution during infection
Functional complementation with rpsC variants lacking ribosomal incorporation ability
Studies in other bacterial pathogens have demonstrated extraribosomal functions for ribosomal proteins, including interactions with host factors and contributions to virulence. Similar principles may apply to V. vulnificus rpsC, particularly given that this organism has demonstrated genomic markers among clinically relevant strains in the form of clinical-associated alleles (CAAs) with potential direct roles in virulence .
Genetic manipulation approaches to study rpsC function in V. vulnificus pathogenicity include:
Conditional expression systems: Since rpsC is likely essential, use of inducible promoters to control expression levels
Domain-specific mutations: Target specific functional domains while preserving ribosomal incorporation
Allelic exchange: Replace native rpsC with variant alleles, particularly testing clinical versus environmental isolate versions
CRISPR interference (CRISPRi): For partial knockdown without complete elimination
Reporter fusions: Create translational or transcriptional fusions to monitor expression patterns during infection
These approaches must be carefully designed to distinguish between effects on general translation and specific virulence functions. A critical experimental design would include complementation controls and careful phenotypic analysis across multiple virulence assays, similar to approaches used in studying RpoS function in V. vulnificus stress response and virulence factor expression .
Potential host factors that may interact with V. vulnificus rpsC during infection include:
Components of the innate immune system:
Pattern recognition receptors
Antimicrobial peptides
Complement proteins
Intracellular factors following invasion:
Cytoskeletal components
Ubiquitination machinery
Autophagy-related proteins
Extracellular matrix components:
Fibronectin
Collagen
Laminin
Identifying these interactions requires:
Pull-down assays using recombinant rpsC with host cell lysates
Yeast two-hybrid or bacterial two-hybrid screening
Protein microarray analysis
Co-immunoprecipitation from infected samples
Understanding these interactions could reveal mechanisms by which V. vulnificus evades host defenses or manipulates host cell functions, contributing to its high pathogenicity and mortality rate in human infections .
V. vulnificus rpsC may contribute to immune evasion through several potential mechanisms:
Molecular mimicry with host proteins to avoid immune recognition
Interference with host signaling pathways to suppress immune responses
Binding and neutralization of host antimicrobial peptides
Modification of outer membrane composition to alter pathogen-associated molecular pattern recognition
These mechanisms can be investigated using:
Infection models with wild-type versus rpsC variant strains
Transcriptomic analysis of host immune response genes during infection
Direct binding assays with immune components
Intracellular survival assays in immune cells
The ability of V. vulnificus to cause rapidly progressing septicemia with high mortality suggests sophisticated immune evasion strategies, potentially involving multiple factors including rpsC.
Detecting rpsC localization during host-pathogen interactions requires sophisticated imaging and biochemical approaches:
Immunofluorescence microscopy:
Fixed cell imaging with rpsC-specific antibodies
Live cell imaging with fluorescently tagged rpsC
Electron microscopy:
Immunogold labeling for transmission electron microscopy
Correlative light and electron microscopy (CLEM)
Biochemical fractionation:
Differential centrifugation of infected host cells
Density gradient separation
Western blot analysis of fractions
Proximity labeling:
BioID or APEX2 fusion proteins
Identification of proximal proteins during infection
These approaches can determine whether rpsC remains exclusively with ribosomes during infection or relocates to other cellular compartments, suggesting additional functions. Similar approaches have been used to study localization patterns of other bacterial factors during infection, providing insights into their multifunctional roles.
V. vulnificus rpsC likely shares core structural features with rpsC proteins from other pathogenic bacteria while possessing species-specific adaptations. Key differences may include:
Sequence variations in surface-exposed regions:
Modifications in RNA-binding domains affecting translation efficiency
Alterations in protein-protein interaction surfaces
Species-specific post-translational modification sites
Functional adaptations:
Differential binding affinities for host factors
Varying contributions to stress responses
Species-specific extraribosomal functions
Comparative analysis techniques include:
Multiple sequence alignment and phylogenetic analysis
Homology modeling based on solved structures
Thermal stability comparisons
Functional complementation studies across species
These differences may contribute to the specific pathogenic profile of V. vulnificus, particularly its ability to cause rapidly progressing septicemia compared to other Vibrio species.
Evolutionary patterns in rpsC across Vibrio species and strains likely show:
Core domains with high conservation reflecting essential ribosomal functions
Hypervariable regions potentially adapting to specific ecological niches
Evidence of horizontal gene transfer and recombination events
Selective pressure signatures in clinical versus environmental isolates
Population genomic approaches can reveal these patterns:
Whole genome sequencing of diverse isolates
Calculation of dN/dS ratios to identify selection pressures
Bayesian evolutionary analysis
Assessment of recombination rates and hotspots
High recombination rates and frequent exchange of genetic elements between V. vulnificus populations have been documented , suggesting that rpsC may show evidence of these evolutionary processes, potentially contributing to the ecological diversification of the species.
Comparisons between clinical and environmental V. vulnificus isolates can provide critical insights for rpsC research:
Identification of clinical-associated alleles (CAAs) in rpsC sequences
Detection of strain-specific post-translational modifications
Correlation between rpsC variants and virulence phenotypes
Understanding environmental adaptations versus host-specific functions
Research methodologies should include:
Comparative genomics across strain collections
Functional characterization of variant proteins
Expression analysis under environmental versus host-mimicking conditions
Virulence assays with isogenic strains differing only in rpsC alleles
This approach aligns with recent findings showing unique genomic markers among clinical V. vulnificus strains in the form of clinical-associated alleles with potential direct roles in virulence , suggesting that similar patterns might be observed in rpsC.
Recombinant V. vulnificus rpsC offers several potential applications in vaccine development research:
As a vaccine candidate:
Recombinant rpsC could serve as a subunit vaccine component
Conserved epitopes may provide cross-protection against multiple strains
Surface-exposed regions could elicit neutralizing antibodies
As an adjuvant:
rpsC may have immunostimulatory properties that enhance vaccine responses
Could be fused to other antigens to improve immunogenicity
For antibody production:
Generate antibodies for diagnostic applications
Develop therapeutic antibodies targeting exposed rpsC
Experimental approaches should include:
Immunization studies with various rpsC formulations
Epitope mapping to identify immunodominant regions
Challenge studies in appropriate animal models
Evaluation of both humoral and cell-mediated responses
Development of vaccine strategies against V. vulnificus is particularly important given its status as a deadly septicemia-causing pathogen with high mortality rates .
When studying rpsC regulation in response to environmental stresses, researchers should consider:
Stress conditions relevant to V. vulnificus ecology and pathogenesis:
Temperature shifts (environmental to host temperature)
Osmotic stress (freshwater to saltwater transitions)
Iron limitation
Acid stress
Oxidative stress
Temporal dynamics:
Immediate responses (0-30 minutes)
Short-term adaptation (1-6 hours)
Long-term responses (12-24+ hours)
Regulatory network analysis:
Methodology considerations:
Use multiple stress application methods
Include appropriate controls for each condition
Consider potential confounding factors
Use time-course measurements rather than single timepoints
This approach mirrors established methods for studying stress responses in V. vulnificus, such as those used to characterize RpoS regulation, where two distinct transcriptional initiation sites were identified and their responses to environmental conditions were characterized .
When investigating rpsC interactions with antibiotics, researchers should follow these experimental design principles:
Antibiotic selection considerations:
Include representatives from different structural classes
Focus on translation-targeting antibiotics
Include both clinically relevant and research compounds
Consider compounds at various development stages
Interaction characterization approaches:
Biochemical binding assays (isothermal titration calorimetry, surface plasmon resonance)
Structural studies (X-ray crystallography, cryo-EM)
Functional impacts (in vitro translation assays)
Resistance development studies
Experimental controls:
Include rpsC proteins from antibiotic-sensitive and resistant strains
Test rpsC variants with site-directed mutations in putative binding sites
Compare with other ribosomal proteins that interact with the same antibiotics
Include appropriate vehicle controls
Translational considerations:
Assess physiological relevance with whole-cell assays
Determine structure-activity relationships
Investigate synergistic effects with other antibiotics
Evaluate resistance mechanisms
Such studies could lead to new therapeutic approaches against V. vulnificus infections, which currently have high mortality rates and require rapid intervention .
Addressing protein degradation issues with recombinant V. vulnificus rpsC requires a comprehensive strategy:
Expression optimization:
Reduce expression temperature to 16-18°C
Use strains lacking key proteases (BL21, Rosetta)
Co-express with chaperones to promote proper folding
Optimize codon usage for expression host
Buffer optimization:
Include protease inhibitor cocktails during all purification steps
Maintain samples at 4°C throughout processing
Add stabilizing agents:
5-10% glycerol
1-5 mM DTT or 2-mercaptoethanol
0.1-0.5 M NaCl to maintain ionic strength
Storage considerations:
Flash-freeze aliquots in liquid nitrogen
Store at -80°C rather than -20°C
Avoid repeated freeze-thaw cycles
Consider lyophilization for long-term storage
Analytical approaches:
Monitor degradation patterns by SDS-PAGE and Western blotting
Use mass spectrometry to identify cleavage sites
Test pH ranges to identify stability optima
Perform thermal shift assays to assess stability
Similar stability challenges have been encountered with other V. vulnificus proteins, requiring careful optimization of expression and purification conditions to maintain structural integrity and function.
Inconsistent results in rpsC-RNA binding assays can be addressed through systematic troubleshooting:
RNA quality considerations:
Ensure RNA is free from degradation (check via denaturing gel electrophoresis)
Verify proper RNA folding using structure probing techniques
Use freshly prepared RNA samples
Consider chemical synthesis for short RNAs versus in vitro transcription for longer constructs
Protein quality factors:
Verify protein activity after each purification
Assess batch-to-batch variation
Test different storage conditions
Determine if post-translational modifications affect binding
Assay optimization:
Standardize buffer components (particularly Mg²⁺ concentration)
Optimize temperature and incubation times
Include appropriate controls for non-specific binding
Test multiple detection methods:
Electrophoretic mobility shift assays (EMSA)
Filter binding assays
Fluorescence anisotropy
Surface plasmon resonance (SPR)
Data analysis improvements:
Use replicate measurements (minimum n=3)
Perform statistical analysis to assess significance
Establish clear criteria for positive binding
Consider cooperative binding models where appropriate
The EMSA approach has been successfully used to study other DNA-protein interactions in V. vulnificus, such as the binding of cAMP-CRP complex to rpoS promoters , and similar principles apply to RNA-protein interaction studies.
When designing in vivo experiments to study rpsC function in V. vulnificus virulence, researchers should consider:
Model system selection:
Mouse models for systemic infection
Cell culture models for specific host-pathogen interactions
Invertebrate models (C. elegans, Galleria mellonella) for initial screening
Ex vivo human tissue models for translational relevance
Genetic manipulation approach:
Conditional expression systems for essential genes
Site-directed mutagenesis targeting specific domains while maintaining ribosomal function
Complementation strategies with variant alleles
Consider dual-function impacts (translation versus extraribosomal roles)
Experimental controls:
Include wild-type parent strain
Use appropriate antibiotic controls
Test multiple bacterial doses
Include time-course measurements
Readout selection:
Survival/mortality assessment
Bacterial burden quantification
Host response measurements (cytokines, histopathology)
Systems biology approaches (transcriptomics, proteomics)
Ethical and statistical considerations:
Ensure proper power calculations for animal studies
Implement appropriate randomization and blinding
Follow the 3Rs principles (replacement, reduction, refinement)
Obtain proper ethical approvals
These approaches align with current practices in V. vulnificus virulence research, which have revealed its ability to cause rapidly progressing septicemia with high mortality rates , necessitating robust in vivo experimental designs to study virulence factors.
CRISPR-based detection systems can be adapted for studying V. vulnificus rpsC variants through several innovative approaches:
SNP detection in rpsC alleles:
Design CRISPR-Cas12a or Cas13a systems targeting allele-specific sequences
Develop multiplexed detection of multiple variants simultaneously
Create rapid genotyping systems for clinical versus environmental alleles
Expression monitoring:
Target rpsC mRNA with Cas13-based detection systems
Quantify expression levels in different conditions
Monitor expression during infection in real-time
Protocol optimization:
Combine with recombinase-aided amplification (RAA) for increased sensitivity
Develop lateral flow assay formats for field applications
Design fluorescent reporter systems for quantitative assessment
The RAA-CRISPR/Cas12a method has already been successfully applied for rapid and sensitive detection of V. vulnificus in 40 minutes with high specificity and sensitivity . Similar approaches could be adapted for rpsC variant detection with appropriate guide RNA design targeting allele-specific sequences.
Advanced structural biology techniques that are particularly informative for studying rpsC ribosomal integration include:
Cryo-electron microscopy (Cryo-EM):
Single-particle analysis of intact ribosomes
Visualization of rpsC in its native ribosomal context
Detection of conformational changes upon ribosome assembly
Resolution now approaching atomic detail (2-3Å)
Integrative structural biology approaches:
Combining X-ray crystallography of individual domains
Small-angle X-ray scattering (SAXS) for solution structure
NMR for dynamic regions
Cross-linking mass spectrometry for interaction networks
Time-resolved structural methods:
Time-resolved cryo-EM with millisecond resolution
Temperature-jump triggered structural transitions
Mixing experiments to capture assembly intermediates
In-cell structural techniques:
Förster resonance energy transfer (FRET) to monitor distances
In-cell NMR to detect structural changes in the cellular environment
Electron tomography of intact cells
These approaches can reveal not only static structural information but also the dynamic process of ribosome assembly and the precise positioning of rpsC within the functional ribosome, providing insights into both canonical translation roles and potential extraribosomal functions.
Systems biology approaches can significantly enhance understanding of rpsC regulation networks in V. vulnificus through:
Multi-omics integration:
Combine transcriptomics, proteomics, and metabolomics data
Correlate rpsC expression with global cellular changes
Identify regulatory hubs controlling rpsC expression
Map post-transcriptional and post-translational modifications
Network analysis techniques:
Construct protein-protein interaction networks
Identify transcription factor binding networks
Model signaling pathways controlling ribosomal protein expression
Perform epistasis analysis to establish regulatory hierarchies
Computational modeling:
Develop mathematical models of rpsC regulation
Perform flux balance analysis to understand metabolic impacts
Create predictive models of regulation under various conditions
Simulate evolutionary trajectories of regulatory networks
Perturbation approaches:
Systematic gene deletion/knockdown studies
Chemical genomics screens
Environmental stress response mapping
Host interaction perturbation analysis
These approaches could reveal how rpsC regulation is integrated with global regulatory networks, similar to how RpoS has been shown to function as a global regulator responding to various stresses and controlling virulence factor expression in V. vulnificus .
The most promising future research directions for V. vulnificus rpsC studies include:
Clinical applications:
Basic science advancements:
Comprehensive characterization of extraribosomal functions
Understanding allelic variations between clinical and environmental isolates
Elucidation of stress-response roles
Mapping of interaction networks within and outside the ribosome
Technological innovations:
Development of rpsC-based biosensors
Creation of engineered rpsC variants with enhanced functions
Application of in vivo structural biology to study dynamics during infection
Ecological and evolutionary studies:
Assessment of rpsC adaptations to environmental niches
Tracking evolutionary trajectories across Vibrio species
Understanding selection pressures on rpsC structure and function
These directions build upon current understanding of V. vulnificus pathogenesis and the dual roles many bacterial proteins play in both basic cellular functions and virulence .
Artificial intelligence and machine learning can advance rpsC research through:
Structural predictions:
Improved protein structure prediction via AlphaFold and RoseTTAFold
Prediction of protein-RNA and protein-protein interactions
Modeling of conformational dynamics
Virtual screening for binding molecules
Functional annotation:
Prediction of functional sites based on conservation patterns
Identification of post-translational modification sites
Classification of variant impact on function
Discovery of cryptic functional domains
Literature mining:
Automated extraction of rpsC-related findings from published literature
Network construction from disparate studies
Hypothesis generation based on literature patterns
Identification of knowledge gaps
Experimental design optimization:
Design of optimal mutagenesis strategies
Prediction of expression conditions for maximum yield
Optimization of buffer compositions
Planning of efficient experimental sequences
These applications could significantly accelerate research progress and provide novel insights into rpsC function that might not be apparent through traditional approaches.
Interdisciplinary collaborations that would most benefit V. vulnificus rpsC research include:
Structural biology and computational chemistry:
High-resolution structure determination
Molecular dynamics simulations
Drug design targeting rpsC-specific features
Modeling of ribosome assembly
Immunology and vaccine development:
Characterization of host immune responses to rpsC
Development of rpsC-based vaccine components
Understanding immune evasion mechanisms
Design of immunodiagnostics
Ecology and evolutionary biology:
Tracking rpsC variations across environmental niches
Understanding selective pressures
Mapping ecological distribution of variants
Correlation with pathogenic potential
Systems biology and bioinformatics:
Integration of multi-omics data
Network analysis of regulatory systems
Comparative genomics across Vibrio species
Predictive modeling of virulence