Recombinant Vibrio vulnificus 60 kDa chaperonin 2 (groL2), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
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 prior arrangement 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 settle 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% and serves as a guideline.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and the protein's inherent 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. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
groL2; groEL2; VVA165960 kDa chaperonin 2; GroEL protein 2; Protein Cpn60 2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Vibrio vulnificus (strain YJ016)
Target Names
groL2
Uniprot No.

Target Background

Function

In conjunction with its co-chaperonin GroES, this protein plays a crucial role in facilitating protein folding. The GroEL-GroES system forms a nano-cage that encapsulates unfolded substrate proteins, providing an optimized environment to promote and accelerate proper folding.

Database Links

KEGG: vvy:VVA1659

Protein Families
Chaperonin (HSP60) family
Subcellular Location
Cytoplasm.

Q&A

What is the role of chaperonin groL2 in Vibrio vulnificus biology?

Chaperonin groL2 in V. vulnificus likely functions as a molecular chaperone that facilitates proper protein folding, particularly under stress conditions. Similar to GroEL proteins in other bacteria, it would form a multi-subunit complex that provides a protected environment for protein folding. In pathogenic bacteria like V. vulnificus, molecular chaperones are often upregulated during infection to counter host-induced stress and maintain proteostasis when transitioning from marine environments to human hosts . This adaptation is particularly relevant given that V. vulnificus causes highly lethal sepsis after consumption of raw oysters .

How does groL2 expression change during infection compared to environmental conditions?

Like other bacterial stress-response proteins, groL2 expression likely increases during host infection when V. vulnificus faces temperature shifts, oxidative stress, and other hostile conditions. Research on V. vulnificus pathogenesis indicates that environmental and clinical strains show distinct genetic and expression patterns . Given that V. vulnificus transitions from oysters (environmental reservoir) to human hosts, groL2 expression would be expected to change significantly during this transition, potentially contributing to the bacterium's ability to establish infection. Experimental approaches to study these expression changes would include qRT-PCR analysis, RNA-seq comparisons between environmental and infection conditions, and reporter gene fusions to monitor expression dynamics in real-time.

What sequence homology exists between V. vulnificus groL2 and chaperonins from other Vibrio species?

While the search results don't directly address sequence homology of groL2, they do mention genetic variation in virulence factors between V. vulnificus and Vibrio anguillarum through recombination events . Researchers studying groL2 should conduct comprehensive sequence alignments using bioinformatics tools like BLAST, Clustal Omega, and phylogenetic analysis to determine conservation levels across Vibrio species. Particular attention should be paid to ATP-binding domains, substrate-binding regions, and oligomerization interfaces, which are functionally critical regions in chaperonins. Identifying unique sequence features in V. vulnificus groL2 could provide insights into species-specific adaptations relevant to its particular environmental niche and pathogenicity.

What structural features distinguish V. vulnificus groL2 from other bacterial chaperonins?

Molecular chaperonins typically share conserved structural elements, but species-specific variations often exist to accommodate particular substrate proteins relevant to the organism's lifestyle. For V. vulnificus, which experiences dramatic environmental transitions between seawater and the human host, structural adaptations in groL2 might include modified substrate-binding regions or altered ATP hydrolysis mechanisms. Researchers should employ X-ray crystallography, cryo-electron microscopy, and molecular dynamics simulations to characterize the three-dimensional structure of recombinant groL2. Particular attention should focus on comparing the apical, intermediate, and equatorial domains with those of related chaperonins to identify structural adaptations that might relate to V. vulnificus pathogenesis .

How do co-chaperonins interact with V. vulnificus groL2 to modulate its function?

Chaperonin systems typically include co-chaperonins (like GroES) that regulate the activity of the main chaperonin complex. In V. vulnificus, understanding the groL2-co-chaperonin interaction requires biochemical approaches including co-immunoprecipitation, isothermal titration calorimetry, and hydrogen-deuterium exchange mass spectrometry. Since V. vulnificus experiences diverse environmental conditions during its lifecycle, the co-chaperonin interaction may have evolved specific regulatory mechanisms that differ from those in other bacteria. Researchers should examine whether environmental signals (like temperature, salinity, or host factors) modify these interactions as part of the bacterial adaptation strategy during infection .

What specific substrate proteins in V. vulnificus depend on groL2 for proper folding?

Given V. vulnificus's potent virulence factors, identifying groL2-dependent substrate proteins could reveal important pathogenicity mechanisms. The search results indicate that V. vulnificus produces several critical virulence factors, including the MARTX(Vv) toxin and VVH (Vibrio vulnificus hemolysin) . Researchers should investigate whether these virulence factors require groL2 for proper folding using approaches such as:

Experimental ApproachTechnical DetailsExpected Outcomes
Co-immunoprecipitationPull-down assays using anti-groL2 antibodies followed by mass spectrometryIdentification of interacting proteins
Proteome analysis in groL2-depleted conditionsConditional knockdown of groL2 combined with quantitative proteomicsDetection of proteins destabilized by groL2 reduction
In vitro folding assaysReconstitution experiments with purified groL2 and candidate substratesDirect demonstration of folding assistance
Genetic suppressor screensIdentification of mutations that rescue groL2 deficiencyDiscovery of functional connections

What are the optimal conditions for expressing and purifying recombinant V. vulnificus groL2?

Producing high-quality recombinant groL2 requires optimization of expression systems and purification protocols. Researchers should consider:

  • Expression system selection: E. coli BL21(DE3) strains are typically suitable for chaperonin expression, though codon optimization may be necessary given potential codon usage differences between E. coli and V. vulnificus.

  • Induction conditions: Lower induction temperatures (16-20°C) often yield better results for large multi-domain proteins like chaperonins, reducing inclusion body formation.

  • Purification strategy: Affinity purification using His-tags followed by size exclusion chromatography to ensure isolation of properly assembled oligomeric complexes.

  • Quality control: Circular dichroism spectroscopy and differential scanning calorimetry to verify proper folding and stability of the purified protein.

  • Activity assays: ATP hydrolysis assays and substrate folding assays to confirm functional activity of the recombinant protein.

This methodological framework is particularly important given the complex oligomeric nature of chaperonin assemblies and their tendency to aggregate when improperly expressed .

How can researchers effectively assess the chaperone activity of recombinant groL2 in vitro?

Assessing chaperone activity requires multiple complementary approaches to comprehensively characterize function:

  • Substrate refolding assays: Using model proteins like malate dehydrogenase or citrate synthase, which become inactivated when denatured and can be assayed for activity recovery after chaperonin-assisted refolding.

  • Aggregation prevention assays: Monitoring light scattering to quantify the ability of groL2 to prevent aggregation of heat-denatured proteins.

  • ATP hydrolysis assays: Measuring ATPase activity, which correlates with the functional cycle of chaperonins, using malachite green or coupled enzymatic assays.

  • Single-molecule FRET studies: Examining conformational changes during the chaperonin functional cycle.

  • Thermal stability enhancement: Assessing the ability of groL2 to increase the thermal stability of client proteins using differential scanning fluorimetry.

These methodologies allow for comprehensive functional characterization beyond simple protein-protein interaction studies .

What genetic approaches are most effective for studying groL2 function in V. vulnificus?

Given the likely essential nature of groL2, researchers should employ sophisticated genetic approaches:

  • Conditional expression systems: Using inducible promoters to control groL2 expression, allowing for depletion studies without completely eliminating this potentially essential gene.

  • Domain-specific mutations: Creating targeted mutations in functional domains (ATP-binding, substrate-binding) rather than complete gene deletions.

  • Complementation studies: Expressing groL2 variants in trans to assess functional conservation across species or to evaluate the impact of specific mutations.

  • Fluorescent tagging: Creating groL2-fluorescent protein fusions to monitor localization and expression patterns during infection processes, particularly during transitions between environmental and host conditions as mentioned in studies of V. vulnificus pathogenesis .

  • CRISPR interference: Using dCas9-based approaches for partial gene repression rather than complete knockout if groL2 proves essential.

These approaches recognize the technical challenges of manipulating potentially essential genes while allowing for functional characterization in the context of V. vulnificus biology .

How might groL2 contribute to V. vulnificus virulence mechanisms?

While the search results don't directly connect groL2 to virulence, they describe several virulence mechanisms that might depend on chaperonin function:

  • Toxin production and assembly: V. vulnificus produces the MARTX(Vv) toxin, which is "an important virulence factor by the intragastric route of infection in mice" . As a large, complex protein, MARTX(Vv) likely requires chaperone assistance for proper folding, particularly during infection when bacteria experience stress conditions.

  • Environmental adaptation: V. vulnificus transitions from oysters to human hosts, encountering temperature shifts, oxidative stress, and immune defenses. GroL2 likely plays a critical role in maintaining proteostasis during these transitions, similar to chaperonins in other pathogens .

  • Stress resistance: The search results mention that V. vulnificus must "overcome the innate immune defenses, including complement-mediated phagocytosis" . Bacterial chaperonins typically contribute to stress resistance by preventing protein misfolding during exposure to host defense mechanisms.

  • Potential immunomodulation: Some bacterial chaperonins are known to have immunomodulatory effects. The search results describe various V. vulnificus interactions with immune cells , suggesting potential roles for groL2 in host-pathogen interactions.

Is there evidence for groL2 involvement in host-pathogen interactions during V. vulnificus infection?

The search results describe multiple V. vulnificus interactions with host cells that might involve groL2:

  • Epithelial cell interactions: V. vulnificus "induces NF-κB-dependent mitochondrial cell death via lipid raft-mediated reactive oxygen species production" . Chaperonins might be involved in maintaining bacterial protein function during this host cell interaction.

  • Immune cell modulation: The search results mention that V. vulnificus interacts with macrophages, lymphocytes, and vascular endothelial cells . Bacterial chaperonins have been reported to be recognized by host immune receptors in other systems, suggesting potential similar roles for groL2.

  • Survival in blood: V. vulnificus causes septicemia, requiring adaptation to the bloodstream environment . GroL2 likely contributes to bacterial survival during this critical phase of infection.

Researchers should investigate these potential interactions using approaches like bacterial two-hybrid screens, pull-down assays with host cell lysates, and immunological studies to determine if groL2 directly interacts with host components.

How does groL2 expression compare between virulent clinical isolates and less pathogenic environmental strains?

The search results highlight differences between clinical and environmental V. vulnificus strains, particularly regarding toxin variants . Similarly, groL2 expression patterns or sequence variations might differ between these strain types:

  • Clinical vs. environmental comparison: Since "most cases occur in patients with underlying conditions" , clinical isolates might show adaptations in groL2 expression or function that enhance survival in compromised hosts.

  • Iron availability response: The results mention that "hepcidin has a critical role in host defense against V. vulnificus by inducing reactive hypoferremia" . GroL2 expression might be differentially regulated in response to iron availability in clinical versus environmental isolates.

  • Growth rate correlation: The search results indicate differences in growth rates between clinical and environmental strains in mouse models . Chaperonin expression often correlates with growth rate, suggesting possible differences in groL2 expression patterns.

Researchers should examine groL2 sequence conservation, expression levels, and functional activity across diverse V. vulnificus isolates to determine if variations correlate with virulence potential.

What potential exists for targeting groL2 in therapeutic development against V. vulnificus infections?

Given the rapid progression and high mortality of V. vulnificus infections, novel therapeutic approaches are needed:

  • Inhibitor development: Researchers should explore small molecule inhibitors specifically targeting V. vulnificus groL2, potentially disrupting its ATPase activity or oligomerization.

  • Combination therapies: Since V. vulnificus produces multiple virulence factors , combining groL2 inhibitors with antibiotics or anti-toxin approaches might yield synergistic effects.

  • Diagnostic applications: If groL2 shows sequence or expression patterns unique to virulent strains, it could serve as a biomarker for rapid identification of high-risk V. vulnificus isolates in seafood or clinical samples.

  • Host-directed therapies: The search results mention that "hepcidin-deficient mouse model of severe hemochromatosis" could be valuable for studying V. vulnificus infections. Therapeutic approaches that combine targeting bacterial groL2 with modulation of host iron metabolism might be particularly effective.

How can systems biology approaches enhance our understanding of groL2's role in V. vulnificus pathogenesis?

Integrative approaches would provide comprehensive insights into groL2 function:

  • Multi-omics integration: Combining transcriptomics, proteomics, and metabolomics data from V. vulnificus under various conditions to identify networks involving groL2.

  • Host-pathogen interaction mapping: Systematic analysis of how groL2 expression correlates with virulence factor production and host response patterns.

  • Ecological modeling: Understanding how groL2 function contributes to V. vulnificus survival in environmental reservoirs like oysters versus during human infection.

  • Evolutionary analysis: Examining how groL2 has evolved in V. vulnificus compared to related species, potentially revealing adaptations specific to its ecological niche and pathogenic lifestyle .

This systems-level understanding could reveal critical intervention points for preventing the "highly lethal sepsis after consumption of raw oysters" that characterizes severe V. vulnificus infections.

What emerging technologies could advance research on V. vulnificus groL2?

Researchers should consider applying cutting-edge approaches to groL2 studies:

  • Cryo-electron microscopy: For high-resolution structural analysis of the entire chaperonin complex under near-native conditions.

  • Single-cell RNA-seq: To examine heterogeneity in groL2 expression within bacterial populations during infection.

  • Advanced mouse models: The search results mention "hepcidin-deficient mouse model" and "NIAAA model...of chronic and binge ethanol feeding" as valuable for studying V. vulnificus. These models could reveal groL2's role in pathogenesis in physiologically relevant settings.

  • CRISPR-based approaches: For precise genetic manipulation to study groL2 function in V. vulnificus.

  • Artificial intelligence-driven protein structure prediction: To model groL2-substrate interactions and identify potential inhibitor binding sites.

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.