Recombinant Vibrio vulnificus Protein sprT (sprT)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder.
Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes if needed. We will fulfill requests whenever possible.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The specific tag type is determined during production. If a particular tag type is required, please inform us, and we will prioritize its inclusion.
Synonyms
sprT; VV2865; Protein SprT
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-168
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Vibrio vulnificus (strain YJ016)
Target Names
sprT
Target Protein Sequence
MKKIEFELHY QAQKALHACI DKANRYFQHQ FPMPLLSYQL RGKAAGKAYL QLNQIRLNPI LFKENKTAFL EEVIPHEVAH LLTYQLYGRV KPHGAEWQSI MQGVFLLPAN TTHQFAVASV QGKTFQYRCQ CREFPLTIRR HNKVLRNEAS YTCQKCRQTL IFTGIQLS
Uniprot No.

Target Background

Database Links

KEGG: vvy:VV2865

Protein Families
SprT family
Subcellular Location
Cytoplasm.

Q&A

What are the major virulence factors in Vibrio vulnificus, and how can they be expressed recombinantly?

V. vulnificus produces several key virulence factors that contribute to its pathogenicity, including:

  • MARTX Vv (Multifunctional-Autoprocessing RTX) toxins with variant effector domains

  • VvsA serine protease (45 kDa chymotrypsin-like alkaline protease)

  • Capsular polysaccharide (CPS)

  • Hemolysins

For recombinant expression, these proteins can be produced using various systems:

Expression SystemAdvantagesLimitationsSuitable for
E. coliHigh yield, cost-effectiveMay form inclusion bodies, lack post-translational modificationsRTX toxin fragments, VvsB
Cell-free translation (RTS)Avoids toxicity issues, rapidLower yield, more expensiveVvsB, potentially toxic proteins
V. vulnificus native expressionNatural folding and modificationsContainment requirements, lower yieldVvsA-VvsB co-expression

For optimal results when expressing V. vulnificus virulence factors, include native operator/promoter (O/P) and Shine-Dalgarno (SD) sequences from the source organism, as studies demonstrate this significantly improves functional protein production .

How does genetic diversity of Vibrio vulnificus impact recombinant protein expression strategies?

V. vulnificus exhibits significant genetic diversity with multiple biotypes and genetic lineages that impact protein expression strategies:

  • Clinical (C-type) and environmental (E-type) genotypic variations exist

  • At least two major phylogenetic lineages (I and II) with different virulence profiles

  • Genetic recombination events create variant toxins with altered potency

When designing recombinant expression, researchers should:

  • Verify the source strain's genotype and lineage classification

  • Consider codon optimization based on the specific strain's genetic preferences

  • Account for potential post-translational modifications unique to the biotype

  • Sequence-confirm the target gene, as significant variants exist even within the same species

The rtxA1 gene, for example, exists in at least four distinct variants encoding toxins with different arrangements of effector domains, requiring tailored expression strategies for each variant .

What expression systems are most effective for producing functional recombinant VvsA and VvsB proteins from Vibrio vulnificus?

Research demonstrates that expression system selection significantly impacts the functionality of V. vulnificus serine protease (VvsA) and its regulatory protein VvsB:

Recommended expression approaches:

  • Rapid Translation System (RTS): For individual expression of VvsB

    • Successfully used to express functionally active VvsB protein

    • Allows study of VvsB's regulatory effects on VvsA activity

  • Native promoter/operator constructs in E. coli: For VvsA

    • Including V. vulnificus-specific O/P and SD sequences significantly enhances activity

    • The pBluescript IIKS+ vector with V. vulnificus regulatory elements shows good results

  • Co-expression considerations:

    • VvsA exhibits higher proteolytic activity when expressed alone compared to co-expression with VvsB

    • This suggests VvsB may suppress VvsA activity intracellularly

Experimental evidence from comparative expression systems:

Expression SystemVvsA ActivityVvsA-VvsB Co-expression ActivityNotes
E. coli with V. vulnificus O/P+SDHighReducedVvsB appears to inhibit activity intracellularly
E. coli without V. vulnificus O/P+SDLowVery lowNative regulatory elements are critical
RTS system for VvsB + native VvsAN/AEnhancedExtracellular addition of purified VvsB (5ng) increases VvsA activity

These findings indicate that VvsB exhibits dual regulatory functions: acting as an inhibitor intracellularly and an activator extracellularly .

What purification strategies minimize degradation of recombinant Vibrio vulnificus proteases while maintaining activity?

Purifying active V. vulnificus proteases presents unique challenges due to their potential for auto-degradation and their sensitivity to inhibitors. Based on research findings, the following strategies are recommended:

  • Rapid single-step affinity purification:

    • Utilize His-tag or GST-tag systems with shortened column contact time

    • Include 10% glycerol in all buffers to stabilize protein structure

    • Maintain cold temperatures (4°C) throughout the purification process

  • Selective inhibitor inclusion during purification:

    • For VvsA purification: Use specific serine protease inhibitors like PMSF (1mM) during cell lysis

    • For MARTX toxins: Avoid metal chelators that might affect toxin structure

  • Compartment-specific extraction protocols:

    • For cytoplasmic fractions: Use gentle lysis with EzBactYeast Crusher kits

    • For periplasmic proteins: Osmotic shock methods yield better results than mechanical disruption

  • Activity preservation considerations:

    • Purify proteins directly into activity-compatible buffers (pH 8.0 for VvsA)

    • When analyzing RtxA1 variants, include protease inhibitor cocktails to prevent degradation by host cell proteases

The choice of expression host also impacts purification success. While E. coli systems are common, they may not provide optimal folding environments for all V. vulnificus proteins .

How can recombinant expression systems be optimized to study the domain-specific functions of MARTX toxin variants from Vibrio vulnificus?

MARTX toxins present particular challenges for recombinant expression due to their large size (~5,200 amino acids) and multiple functional domains. Advanced strategies include:

  • Domain-specific expression approaches:

    • Express individual effector domains separately to study specific functions

    • Use toxin fragments containing the cysteine protease domain (CPD) to study autoprocessing

    • Create chimeric constructs between different MARTX variants to map determinants of potency

  • Inducible expression systems with tight regulation:

    • Use pBAD or T7lac-based systems to minimize toxicity during bacterial growth

    • Employ split-protein complementation to reconstitute toxic activities only after purification

    • Consider cell-free translation for highly toxic domains

  • Structure-guided construct design:

    • The MARTX Vv toxin contains conserved repeat regions at N and C termini essential for secretion

    • Central region includes the cysteine protease domain (CPD) required for inositol hexakisphosphate-induced autoprocessing

    • Design constructs that preserve critical cleavage sites at leucine residues between effector domains

  • Analytical approaches to verify domain functionality:

    • Monitor autoprocessing via size-shift assays with/without inositol hexakisphosphate

    • Assess domain-specific activities: actin depolymerization, ROS induction, caspase-1 activation

    • Analyze membrane binding capacity of N/C-terminal repeat regions

Research has identified four distinct MARTX Vv variants (types A, B, C, and M), each requiring tailored expression strategies to preserve their unique arrangements of effector domains .

What are the methodological approaches for analyzing how VvsB regulates VvsA activity in both intracellular and extracellular environments?

Recent research has revealed a complex dual regulatory role of VvsB on VvsA serine protease activity that differs between intracellular and extracellular environments. To study this phenomenon:

  • Comparative activity assays:

    • Express VvsA alone and measure baseline proteolytic activity

    • Co-express VvsA and VvsB and compare activity levels

    • Purify VvsB separately and add to VvsA-containing culture supernatants at varying concentrations (1ng to 1μg)

    • Monitor elastolytic activity using elastin-Congo red substrate

  • Protein-protein interaction studies:

    • Employ co-immunoprecipitation with tagged variants of VvsA and VvsB

    • Use surface plasmon resonance to measure binding kinetics under different pH/salt conditions

    • Perform cross-linking experiments followed by mass spectrometry to identify interaction sites

  • Structural biology approaches:

    • Conduct hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

    • Consider cryo-EM or X-ray crystallography of the VvsA-VvsB complex

    • Use site-directed mutagenesis to create interaction-deficient variants

  • Compartment-specific activity monitoring:

    • Create fluorescent protein fusions to track localization and activity simultaneously

    • Employ subcellular fractionation to isolate cytoplasmic, periplasmic, and secreted fractions

    • Design cell-based assays that distinguish between internal and external protease activities

Key experimental findings and recommendations:

VvsB ConcentrationEffect on Extracellular VvsA ActivityMethodological Approach
5ngMaximum enhancement (optimal)Add purified VvsB to culture supernatant
10-100ngSlight decrease in enhancementTitration experiments are essential
>1μgNo additional benefitMonitor concentration-dependent effects

Research indicates VvsB inhibits VvsA activity intracellularly to prevent autolysis but facilitates VvsA activation in the extracellular environment—this complex regulation likely contributes to V. vulnificus environmental adaptability .

How can transcriptome analysis be integrated with recombinant protein studies to understand the regulation of virulence factor expression in Vibrio vulnificus?

Integrating transcriptome analysis with recombinant protein studies provides powerful insights into virulence regulation mechanisms:

  • Dual-transcriptome sequencing approaches:

    • Sequence both pathogen and host transcriptomes simultaneously during infection

    • Compare expression profiles in serum versus environmental conditions

    • Identify key regulators like RpoS that control virulence gene expression

  • Transcription factor reconstitution assays:

    • Recombinantly express transcription factors identified in transcriptome studies

    • Perform in vitro transcription assays with purified factors and promoter regions

    • Establish reporter systems to validate regulatory networks in vivo

  • Stress response pathway reconstruction:

    • Express components of the stressosome (RsbR, RsbS, RsbT) identified in transcriptome analyses

    • Establish in vitro phosphorylation cascades with purified proteins

    • Create transcription factor binding assays to map regulatory interactions

  • Condition-specific experimental design:

    • Create exposure conditions that mimic host environments (human serum at 37°C)

    • Contrast with environmental conditions (artificial seawater at 22°C)

    • Monitor temporal expression changes to capture dynamic regulation

Research-proven approaches with demonstrated results:

ConditionRegulatory SystemUpregulated Virulence FactorsExperimental Approach
Human serum (37°C)RpoH (heat shock sigma factor)MARTX toxins, hemolysins, proteasesRNA-seq followed by qRT-PCR validation
Seawater (22°C)RpoS, RpoN, IscRAdherence factors, biofilm componentsMembrane diffusion chambers with natural estuarine waters
Host cell contactRpoSRtxA1 toxin, RtxB1 transporterCo-culture with HeLa cells followed by Western blotting

Transcriptome studies reveal that V. vulnificus employs cyclic-di-GMP signaling to orchestrate a dichotomous genetic switch between "virulence" and "environmental" profiles, which has significant implications for recombinant expression strategies .

What are the common issues encountered when expressing recombinant Vibrio vulnificus toxins, and how can they be addressed?

Researchers frequently encounter several challenges when expressing V. vulnificus toxins recombinantly:

  • Low expression levels or toxicity to host cells:

    • Solution: Use tightly controlled inducible promoters (e.g., arabinose-inducible systems)

    • Solution: Express non-toxic subdomains or use inactive mutants (e.g., for RtxA1)

    • Solution: Lower induction temperature to 16-20°C to reduce toxicity

  • Improper folding and loss of activity:

    • Solution: Include V. vulnificus native operator/promoter and SD sequences

    • Solution: Co-express with bacterial chaperones (GroEL/ES, DnaK/J)

    • Solution: For VvsA/B, expression yields higher activity with V. vulnificus-specific regulatory elements

  • Premature degradation during expression/purification:

    • Solution: Use protease-deficient host strains (e.g., BL21)

    • Solution: Include appropriate protease inhibitors during purification

    • Solution: Add stabilizing agents like glycerol (10%) and reducing agents

  • Lack of post-translational modifications:

    • Solution: Consider expression in eukaryotic systems for complex modifications

    • Solution: Identify minimal modification requirements through domain mapping

    • Solution: For VvsA, proper activation may require VvsB accessory protein

Comparative success rates with different expression strategies:

Virulence FactorExpression ChallengeSuccessful StrategyActivity Recovery
MARTX Vv toxinHost toxicityDomain-specific expressionUp to 70% of native activity
VvsA serine proteaseImproper processingInclude native regulatory elements>80% with O/P and SD sequences
RtxA1 variantsDegradation during purificationProtease inhibitor cocktailsVariant-dependent (40-90%)

For the MARTX toxin, expression of smaller functional domains (400-600 amino acids) has proven more successful than attempting to express the full 5,206-amino acid protein .

How can researchers address contradictory findings regarding the potency of different RtxA1 toxin variants in experimental systems?

Contradictory findings regarding RtxA1 toxin variants present significant challenges for researchers. These can be systematically addressed through:

  • Standardized expression and purification protocols:

    • Use identical expression systems across variant comparisons

    • Implement consistent purification strategies with equivalent buffer conditions

    • Quantify protein concentration through multiple methods (Bradford, BCA, and amino acid analysis)

  • Controlled activity assays:

    • Develop dose-response curves for each toxin variant

    • Standardize cell types and culture conditions for cytotoxicity assays

    • Include multiple readouts of toxicity (LDH release, ATP depletion, morphological changes)

  • Genetic background considerations:

    • Account for the genomic context of different isolates

    • Recognize that toxin potency may depend on other virulence factors

    • Consider lineage-specific effects (lineage I vs. lineage II strains)

  • Resolution strategies for specific contradictions:

    • The unexpected finding that clinical isolates often carry lower-potency M-type toxins despite causing severe disease

    • The observation that environmental potency may not correlate with human virulence

    • Discrepancies between in vitro and in vivo toxin effects

Research approach to resolve contradictions:

  • Isolate each RtxA1 variant and express under identical conditions

  • Perform side-by-side potency testing in multiple cell types

  • Conduct domain-swapping experiments to identify potency determinants

  • Test variants in both intragastric and wound infection mouse models

What emerging technologies could advance the structural and functional characterization of recombinant Vibrio vulnificus virulence factors?

Several cutting-edge technologies offer promising approaches for deeper characterization of V. vulnificus virulence factors:

  • Cryo-EM for large toxin complexes:

    • Enables visualization of full-length MARTX toxins (>5,000 amino acids)

    • Can capture dynamic states during autoprocessing and membrane interaction

    • Allows study of toxin-receptor complexes in near-native conditions

  • Single-molecule biophysics:

    • Utilizes optical tweezers to study mechanical properties of toxin-membrane interactions

    • Employs single-molecule FRET to track conformational changes during activation

    • Enables real-time observation of toxin translocation processes

  • Mass spectrometry proteomics:

    • Hydrogen-deuterium exchange MS to map protein-protein interaction interfaces

    • Cross-linking MS to capture transient VvsA-VvsB interactions

    • Targeted proteomics to quantify effector domain release from MARTX toxins

  • Advanced genetic approaches:

    • CRISPR-based genome editing to create specific domain variants in native context

    • Base editors for precise amino acid substitutions in large toxin genes

    • Biosynthetic gene clusters for producing toxin variants with non-natural amino acids

  • Microfluidic systems with live-cell imaging:

    • Real-time tracking of toxin activity in host cells using fluorescent biosensors

    • Controlled exposure systems to simulate host-pathogen environmental transitions

    • Single-cell analysis of toxin susceptibility and resistance mechanisms

These technologies would address key knowledge gaps, including full-length toxin structures, dynamics of VvsB regulation of VvsA, and the mechanism of RtxA1 effector domain delivery into host cells .

How might understanding genetic recombination in Vibrio vulnificus rtxA1 inform strategies to detect or counter emerging hypervirulent strains?

Research on rtxA1 genetic recombination reveals ongoing evolution that could lead to emergence of novel hypervirulent strains:

  • Surveillance and early detection approaches:

    • Develop PCR-based assays targeting known recombination hotspots in rtxA1

    • Create rapid sequencing workflows to detect novel domain arrangements

    • Implement metagenomic surveillance of environmental samples to track variant prevalence

  • Structure-function prediction models:

    • Utilize machine learning to predict virulence potential of novel domain combinations

    • Create structure-based computational models to assess toxin potency

    • Develop domain-specific antibody arrays for rapid toxin typing

  • Preventative countermeasures:

    • Design broad-spectrum inhibitors targeting conserved toxin regions

    • Develop therapeutic antibodies against common effector domains

    • Create vaccines based on conserved toxin epitopes with cross-protection potential

  • Risk assessment frameworks:

    • Establish databases correlating domain arrangements with clinical outcomes

    • Create predictive models for environmental conditions favoring hypervirulent variants

    • Develop standardized potency assays for comparative toxin evaluation

Research evidence for ongoing recombination and potential emergence:

Recombination SourceResulting VariantClinical SignificanceDetection Method
Plasmid-borne rtxA genesACD+ variants (O-type)Altered cytotoxicity mechanismsPCR targeting ACD domain
V. anguillarum rtxAD-type variantsModified host specificityWhole-genome sequencing
Novel environmental sourcesPotential future variantsUnknown, potentially increased virulenceMetagenomic surveillance

The identification of at least four distinct rtxA1 variants (with different arrangements of effector domains) demonstrates that this toxin undergoes significant genetic rearrangement and may acquire novel domains that could increase virulence potential in the future .

What are the potential applications of recombinant Vibrio vulnificus proteins in biotechnology and medical research?

Despite their pathogenic origins, recombinant V. vulnificus proteins offer several promising applications:

  • Therapeutic enzyme development:

    • VvsA serine protease as a debridement agent for necrotic tissue

    • Modified RtxA1 domains as targeted cell delivery vehicles

    • Engineered toxin variants as cancer cell-specific cytotoxic agents

  • Diagnostic tool development:

    • Recombinant toxin domains as positive controls in diagnostic assays

    • Antibodies against variant-specific domains for strain typing

    • Biosensors utilizing toxin-receptor interactions for environmental monitoring

  • Basic research applications:

    • MARTX toxin domains as tools to study cellular processes:

      • Actin cytoskeleton dynamics

      • Programmed cell death pathways

      • Intracellular signaling mechanisms

  • Protein engineering platforms:

    • VvsA/VvsB as a model system for studying protease regulation

    • MARTX autoprocessing domains as controlled protein cleavage tools

    • Toxin secretion systems as models for protein export technologies

  • Vaccine development:

    • Detoxified recombinant toxin domains as vaccine candidates

    • Multicomponent vaccines incorporating multiple virulence factors

    • Reverse vaccinology approaches using whole proteome analysis

The multifunctional nature of these proteins, particularly the modular structure of MARTX toxins with distinct effector domains and the sophisticated regulation of VvsA by VvsB, provides unique opportunities for biotechnological applications beyond the study of pathogenesis .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.