Recombinant Chromobacterium violaceum ATP-dependent protease subunit HslV (hslV)

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 during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard 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 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
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag type, please inform us; we will prioritize its development.
Synonyms
hslV; CV_0401; ATP-dependent protease subunit HslV; EC 3.4.25.2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-183
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Chromobacterium violaceum (strain ATCC 12472 / DSM 30191 / JCM 1249 / NBRC 12614 / NCIMB 9131 / NCTC 9757)
Target Names
hslV
Target Protein Sequence
MQQFDGTTIV SVRRGERVAL GGDGQVTLGN IVIKATARKI RKLHGGKVLA GFAGGTADAF TLIERFEAKL QKHQGNLLVS AVELAKDWRT DRMLRRLEAM LIVADKDHTL IITGNGDVLE PEQGIAAIGS GGAFAQSAAR ALFENTDLAP EVVVKKSLEI AGDICIYTNH NHLIETLGPD DEA
Uniprot No.

Target Background

Function
Protease subunit of a proteasome-like degradation complex; a key component of the general cellular protein degradation machinery.
Database Links

KEGG: cvi:CV_0401

STRING: 243365.CV_0401

Protein Families
Peptidase T1B family, HslV subfamily
Subcellular Location
Cytoplasm.

Q&A

What is Chromobacterium violaceum and why is its HslV protease of research interest?

Chromobacterium violaceum is a Gram-negative betaproteobacterium abundant in soil and water microbiota of tropical and subtropical regions worldwide. It has emerged as an important model of an environmental opportunistic pathogen with high virulence in human infections . The HslV protease subunit in C. violaceum is of particular research interest because it represents a bacterial homolog of the eukaryotic proteasome beta subunits, functioning as part of an ATP-dependent proteolytic complex essential for protein quality control and stress response. Similar to the well-characterized E. coli counterpart, C. violaceum HslV likely associates with HslU (an ATPase) to form a functional proteolytic complex involved in degrading misfolded or damaged proteins . Understanding this system provides insights into bacterial protein degradation mechanisms and potentially into virulence factors that may contribute to C. violaceum's pathogenicity.

What are the structural characteristics of HslV in C. violaceum compared to other bacterial species?

HslV in C. violaceum shares structural similarities with other bacterial HslV proteins, particularly the well-characterized 19-kDa HslV from E. coli, which functions as a proteasome-like component . Based on conservation across bacterial species, C. violaceum HslV likely forms a ring-shaped oligomeric structure that associates with the ATPase component HslU. Electron micrographs of HslV-HslU complexes reveal ring-shaped particles similar to en face images of the 20S proteasome or the ClpAP protease . Unlike eukaryotic proteasomes, the bacterial HslV proteases (including that from C. violaceum) typically lack tryptic-like and peptidyl-glutamyl-peptidase activities, suggesting a more specialized catalytic function . The active site likely contains a catalytic threonine residue, as HslV activity is inhibited by specific proteasome inhibitors but not by inhibitors of other classes of proteases.

How does the heat shock response regulate HslV expression in C. violaceum?

The hslV gene in bacteria, including C. violaceum, is typically part of a heat shock locus (hsl) that is upregulated during stress conditions. Similar to E. coli, where the activity increased 10-fold in cells expressing heat-shock proteins constitutively , C. violaceum likely exhibits increased expression of HslV under stress conditions. The regulation probably occurs through sigma factors dedicated to heat shock response, such as σ32 (RpoH) in related bacteria. This response ensures that protein quality control mechanisms are enhanced during stress conditions to manage increased levels of misfolded proteins. While specific data on C. violaceum's heat shock regulation is limited, the conservation of heat shock response mechanisms across bacteria suggests similar regulatory pathways control HslV expression in this organism.

What are the optimal expression systems for producing recombinant C. violaceum HslV?

For optimal expression of recombinant C. violaceum HslV, E. coli-based expression systems are typically most effective due to well-established protocols and high yield potential. Expression vectors containing strong inducible promoters such as T7 (pET system) or tac provide controlled expression, with best results often achieved using E. coli BL21(DE3) or derivatives that lack key proteases. Expression should be optimized at lower temperatures (16-25°C) following induction to enhance proper folding of the protease subunit.

The expression vector should include:

  • The complete hslV gene sequence from C. violaceum

  • An appropriate affinity tag (His6 is commonly used) preferably at the N-terminus

  • A precision protease cleavage site for tag removal if necessary for activity studies

For functional studies of the HslV-HslU complex, co-expression strategies using dual-compatible plasmids or bicistronic constructs containing both hslV and hslU genes have proven effective in related systems . This approach increases the likelihood of isolating the functional protease complex rather than individual subunits.

What purification strategies yield the highest activity for recombinant HslV?

A multi-step purification strategy is recommended to obtain highly active recombinant HslV protein:

  • Initial Capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA or similar resins for His-tagged HslV, with careful optimization of imidazole concentrations in wash and elution buffers (typically 20-40 mM for washing, 250-300 mM for elution).

  • Intermediate Purification: Ion exchange chromatography (typically Q-Sepharose) at pH 8.0 to separate HslV from contaminants with different charge properties.

  • Polishing Step: Size exclusion chromatography using Superdex 200 columns to isolate properly assembled HslV oligomers and remove aggregates.

Throughout purification, include ATP (1-2 mM) in buffers when isolating the HslV-HslU complex, as ATP significantly enhances complex stability and activity. For the highest purity, consider:

  • Maintaining reducing conditions with 1-5 mM DTT or 0.5-2 mM TCEP

  • Using buffers containing 50-100 mM salt to prevent non-specific interactions

  • Keeping glycerol concentration at 10-15% to enhance protein stability

  • Performing purification at 4°C to minimize proteolytic degradation

The final purified HslV should be assessed using peptidase activity assays with fluorogenic substrates such as Z-Gly-Gly-Leu-AMC, where activity increases up to 150-fold in the presence of ATP .

How can the functional integrity of purified recombinant HslV be verified?

The functional integrity of purified recombinant HslV from C. violaceum can be verified through multiple complementary approaches:

  • Peptidase Activity Assays: The primary functional test involves measuring hydrolysis of fluorogenic peptide substrates such as Z-Gly-Gly-Leu-AMC in the presence of ATP and the HslU component. A 150-fold ATP-dependent stimulation of activity should be observed for properly folded and assembled complexes . Importantly, other nucleoside triphosphates, non-hydrolyzable ATP analogs, ADP, or AMP should not significantly stimulate activity.

  • Inhibitor Sensitivity Profile: Test inhibition patterns using:

    • Proteasome inhibitors (should inhibit activity)

    • Serine protease inhibitors (should have limited effect)

    • Cysteine, aspartic, and metalloprotease inhibitors (should not inhibit)

  • Complex Formation Analysis:

    • Native PAGE to verify the formation of higher-order HslV-HslU complexes

    • Size exclusion chromatography to confirm proper oligomer assembly

    • Electron microscopy to visualize ring-shaped particles characteristic of functional complexes

  • Thermal Stability Assessment:

    • Differential scanning fluorimetry to determine melting temperature

    • Activity retention after incubation at different temperatures (42-45°C)

  • ATP Hydrolysis Coupling:

    • Measure ATP hydrolysis rates when HslV is combined with HslU

    • Verify that peptide degradation correlates with ATP consumption

A properly functional HslV should demonstrate significant enhancement of proteolytic activity when combined with HslU and ATP, with characteristic inhibition patterns matching those observed for proteasome-like proteases rather than other protease classes.

What are the methodological approaches for studying HslV interactions with potential protein substrates?

Researchers investigating HslV interactions with potential protein substrates can employ several methodological approaches:

1. In vitro Degradation Assays:

  • Purify candidate substrate proteins and incubate with reconstituted HslV-HslU complex

  • Monitor degradation by SDS-PAGE, western blotting, or using fluorescently labeled substrates

  • Compare degradation rates in the presence/absence of ATP to confirm specificity

  • Use mass spectrometry to identify cleavage sites and degradation products

2. Substrate Trapping Approaches:

  • Generate catalytically inactive HslV variants (e.g., through mutation of the catalytic threonine)

  • Use these variants to trap substrates that bind but cannot be degraded

  • Perform pull-down assays followed by mass spectrometry to identify interacting proteins

  • Cross-linking studies to capture transient interactions between HslV and substrates

3. Proteomics-Based Methods:

  • Compare protein profiles between wild-type and ΔhslV C. violaceum strains

  • Use stable isotope labeling (SILAC) to quantify protein turnover rates

  • Apply global proteomics to identify proteins that accumulate in HslV-deficient cells

4. Genetic Approaches:

  • Construct C. violaceum strains with inducible or repressible hslV expression

  • Create bacterial two-hybrid systems to screen for HslV-interacting proteins

  • Use transposon mutagenesis to identify genetic interactions with hslV

5. Structural Biology Methods:

  • Cryo-electron microscopy to visualize substrate engagement by the HslV-HslU complex

  • X-ray crystallography to determine structures of HslV bound to model substrates or inhibitors

  • NMR studies to map the binding interface between HslV and substrate proteins

These approaches can be integrated to build a comprehensive understanding of HslV substrate recognition and processing in C. violaceum.

How can researchers effectively engineer HslV variants with modified substrate specificity?

Engineering HslV variants with modified substrate specificity requires strategic modifications based on structural knowledge and systematic characterization:

Rational Design Approaches:

  • Active Site Modification: Target residues lining the substrate-binding pocket based on homology modeling with E. coli HslV or crystallographic data

  • Entrance Channel Engineering: Modify residues at the pore entrance to alter size selectivity for substrates

  • Interface Mutations: Alter the HslV-HslU interface to modify how substrate translocation occurs

Experimental Protocol for HslV Engineering:

  • Site-Directed Mutagenesis:

    • Create a library of variants with single or combined mutations in the active site

    • Focus on residues corresponding to those determining specificity in other prokaryotic proteases

    • Introduce mutations that mimic specificities of other proteases (e.g., ClpP, Lon)

  • High-Throughput Screening:

    • Develop fluorogenic peptide libraries with diverse sequences

    • Screen variants against these libraries to identify altered cleavage preferences

    • Quantify changes in specificity constants (kcat/Km) for different substrates

  • Validation With Protein Substrates:

    • Test promising variants against model protein substrates

    • Use degradation kinetics to confirm altered specificity

    • Perform competition assays between wild-type and modified substrates

  • Structural Confirmation:

    • Determine crystal structures of engineered variants

    • Use computational docking to predict interactions with new substrates

    • Verify structural integrity of the modified HslV oligomer

Table 1: Examples of Residue Positions for Targeted Mutagenesis in HslV

Position TypeFunctional RoleMutagenesis StrategyExpected Effect
S1 PocketDetermines primary specificityConservative substitutionsAltered preference for P1 residue
Catalytic ThrNucleophile for peptide bond cleavageT→S substitutionMaintained but reduced activity
Entrance LoopControls substrate accessSize/charge alterationsModified substrate selection
HslU InterfaceMediates ATP-dependent activationConservative changesAltered regulation of activity
Oligomerization InterfaceMaintains ring structureStabilizing mutationsEnhanced complex stability

Successful engineering requires iterative optimization and comprehensive characterization of each variant's catalytic parameters, structural integrity, and substrate preference profiles.

What are the optimal assay conditions for measuring C. violaceum HslV proteolytic activity?

Optimized Assay Conditions for C. violaceum HslV Activity:

Buffer Composition:

  • 50 mM Tris-HCl (pH 8.0)

  • 100 mM KCl

  • 5 mM MgCl₂

  • 1 mM DTT

  • 0.5 mg/ml BSA (to prevent surface adsorption)

  • 10% glycerol (for stability)

Essential Components:

  • Purified HslV (0.1-0.5 μM)

  • Purified HslU (0.5-1.0 μM, typically in 2:1 ratio with HslV)

  • ATP (2 mM freshly prepared)

  • Fluorogenic peptide substrate (Z-Gly-Gly-Leu-AMC, 50-100 μM)

Reaction Conditions:

  • Temperature: 30-37°C (optimum typically 37°C)

  • Time course: Continuous monitoring for 30-60 minutes

  • Excitation/emission for AMC detection: 380 nm/460 nm

Control Reactions:

  • No ATP control (essential baseline)

  • ATP-regenerating system (2 mM phosphoenolpyruvate, 2 U/ml pyruvate kinase) for extended assays

  • Substitution of ATP with other nucleotides (ADP, GTP, non-hydrolyzable ATP analogs) should show minimal activity

Activity Calculation:
Calculate specific activity as nmol AMC released per minute per mg of HslV protein. ATP-dependent activity should show 150-fold enhancement over basal activity .

Validation Criteria:

  • Linear reaction kinetics during the measurement period

  • Proportional activity with enzyme concentration

  • Inhibition by proteasome inhibitors but not by other protease inhibitors

  • No significant activity with heat-inactivated enzyme

Alternative substrates such as casein-FITC or FITC-labeled peptides can provide complementary data but typically show lower sensitivity than the Z-Gly-Gly-Leu-AMC substrate.

How can researchers troubleshoot common problems with recombinant HslV expression and activity?

Troubleshooting Guide for Recombinant HslV Production and Activity:

Problem 1: Poor Expression Yield

Potential CauseDiagnostic ApproachSolution Strategy
Toxicity to host cellsGrowth curve analysis after inductionReduce induction temperature (16-20°C), use tightly regulated promoters
Codon biasAnalyze rare codons in hslV sequenceUse Rosetta or similar strain with rare tRNA supplementation
Protein instabilityWestern blot analysis at different times post-inductionCo-express chaperones (GroEL/ES), add protease inhibitors
Insoluble inclusion bodiesAnalyze soluble vs. insoluble fractionsLower IPTG concentration (0.1-0.2 mM), reduce growth temperature

Problem 2: Lack of Proteolytic Activity

Potential CauseDiagnostic ApproachSolution Strategy
Inactive HslU componentTest ATP hydrolysis by HslUEnsure proper folding of HslU, verify ATPase activity
Improper complex assemblyNative PAGE or size exclusion analysisOptimize salt conditions (50-200 mM KCl) to promote complex formation
Oxidative damageActivity with/without reducing agentsInclude 1-5 mM DTT in all buffers
Incorrect HslV:HslU ratioTitration of componentsTest different ratios (1:1, 1:2, 1:3) of HslV:HslU
Inhibitory contaminantsActivity after additional purificationAdd additional purification step (e.g., ion exchange)

Problem 3: Inconsistent Activity Measurements

Potential CauseDiagnostic ApproachSolution Strategy
ATP degradationCompare fresh vs. stored ATP solutionsPrepare ATP fresh or use ATP-regenerating system
Substrate variabilityTest multiple substrate batchesUse internal standards, prepare larger single substrate batches
Temperature fluctuationsMonitor reaction temperatureUse temperature-controlled plate reader or water bath
Protein aggregationDynamic light scatteringAdd stabilizers (glycerol, low concentrations of detergent)
Buffer component interferenceSystematic buffer variationTest minimal buffer systems, eliminate potentially interfering components

General Recovery Strategies:

  • Refolding protocols using stepwise dialysis if inclusion bodies form

  • Buffer optimization screening with varying pH (7.0-8.5) and salt (50-300 mM)

  • Addition of molecular crowding agents (PEG 3350, 2-5%) to stabilize complexes

  • Co-expression with bacterial chaperone systems to improve folding

What are the best approaches for studying HslV in the context of C. violaceum pathogenicity?

Research Approaches for Studying HslV in C. violaceum Pathogenicity:

1. Genetic Manipulation Strategies:

  • Generate clean deletion mutants (ΔhslV) using allelic exchange techniques

  • Create complemented strains with wild-type and mutant variants

  • Develop conditional expression systems (inducible/repressible) to study HslV depletion effects

  • Engineer reporter fusions (hslV-gfp) to monitor expression during infection

2. Infection Model Systems:

  • Establish cell culture models using hepatocytes and macrophages (key target cells)

  • Implement C. elegans infection models for whole-organism studies

  • Utilize murine infection models focusing on liver colonization and clearance

  • Monitor bacterial survival in human serum to assess complement resistance

3. Host-Pathogen Interaction Analyses:

  • Study inflammasome activation in response to C. violaceum with wild-type or ΔhslV strains

  • Examine role of HslV in intracellular survival within professional phagocytes

  • Investigate connections between HslV function and T3SS activity, as T3SS is critical for virulence

  • Assess impact of HslV on host cell pyroptosis, a key clearance mechanism

4. Stress Response Connection:

  • Determine survival rates of wild-type versus ΔhslV strains under conditions mimicking host environments:

    • Oxidative stress (H₂O₂, peroxynitrite)

    • Nutrient limitation

    • Antimicrobial peptide exposure

    • Temperature stress (37°C vs. environmental temperature)

5. Proteomic Approaches:

  • Compare virulence factor production between wild-type and ΔhslV strains

  • Identify differentially regulated proteins during infection using quantitative proteomics

  • Determine if HslV affects the composition or function of bacterial secretomes

  • Monitor proteome changes in response to host defense mechanisms

6. Integration with Quorum Sensing:

  • Investigate potential links between HslV function and the CviI/R quorum sensing system

  • Test if violacein production, regulated by quorum sensing , is affected by HslV activity

  • Examine if HslV processes regulatory factors involved in quorum sensing cascades

These approaches provide complementary data on how HslV contributes to C. violaceum pathogenicity, potentially revealing new therapeutic targets for this emerging opportunistic pathogen.

How does C. violaceum HslV compare functionally to the 20S proteasome and other ATP-dependent proteases?

C. violaceum HslV represents a fascinating evolutionary link between simpler bacterial proteases and the more complex eukaryotic proteasome system. Key comparative aspects include:

Structural Comparison:
HslV forms a simpler proteolytic core compared to the eukaryotic 20S proteasome, likely consisting of two hexameric rings of HslV subunits (12 subunits total) versus the 28-subunit structure (α₇β₇β₇α₇) of the 20S proteasome. Electron micrographs reveal ring-shaped particles similar to en face images of the 20S proteasome or ClpAP protease . Unlike the 20S proteasome, which requires the 19S regulatory particle for ATP-dependent activities, HslV directly associates with its ATPase component HslU.

ATP Dependence and Regulation:
The HslV-HslU system shows stringent ATP dependence, with activity increasing up to 150-fold in the presence of ATP, while other nucleotides have no effect . This specificity parallels the ATP requirement of the 26S proteasome but differs from some other bacterial ATP-dependent proteases like Lon, which can exhibit ATP-independent peptidase activity.

Evolutionary Significance:
HslV represents a simpler ancestral form of the proteasome, providing insights into proteasome evolution. While eukaryotes evolved the more complex 20S proteasome, many bacteria maintained the simpler HslV-HslU system, suggesting this arrangement is optimal for bacterial physiology and environmental adaptation.

Table 2: Functional Comparison of ATP-Dependent Proteases

FeatureC. violaceum HslV-HslU20S/26S ProteasomeClpAP/ClpXPLon
Proteolytic CoreHslV (threonine protease)β subunits (threonine protease)ClpP (serine protease)Serine protease domain
ATPase ComponentHslU19S regulatory particleClpA/ClpXIntegrated ATPase domain
Oligomeric StructureDouble hexameric rings + HslUFour heptameric rings (α₇β₇β₇α₇)Heptameric ringsHexameric rings
Substrate SpecificityChymotrypsin-likeChymotrypsin-like, trypsin-like, caspase-likeBroadBroad
ATP DependenceAbsolute (150-fold activation)AbsoluteAbsolutePartial

What evolutionary insights can be gained from studying HslV across Chromobacterium species?

Studying HslV across Chromobacterium species offers valuable evolutionary insights into bacterial proteolytic systems and their adaptation to diverse ecological niches:

Conservation and Divergence Patterns:
Analysis of HslV sequences across Chromobacterium species reveals patterns of conservation in catalytic residues and substrate-binding regions, while allowing identification of species-specific variations that may reflect ecological adaptations. Species with different lifestyles (free-living versus host-associated) may show distinct HslV sequence features or regulatory patterns. Similar to how the Cpi-1 T3SS is widespread among Chromobacterium species , HslV is likely broadly conserved but with subtle variations reflecting evolutionary pressures.

Coevolution with Virulence Determinants:
Examination of how HslV has coevolved with virulence factors across Chromobacterium species can reveal functional relationships. For instance, species with more complex virulence mechanisms may show corresponding adaptations in their protein quality control systems, including HslV. The widespread occurrence of virulence determinants like T3SS across the Chromobacterium genus suggests that associated protein quality control systems may show parallel evolutionary patterns.

Environmental Adaptation Signatures:
Chromobacterium species occupy diverse environmental niches ranging from soil and water to plant roots, with some capable of causing human infections. Comparative analysis of HslV from these diverse isolates can reveal signatures of adaptation to different stress conditions:

  • Thermal stress adaptations in species from variable temperature environments

  • Oxidative stress resistance features in species that interact with host immune systems

  • Acid tolerance adaptations in species that must survive gastric passage

Horizontal Gene Transfer Assessment:
Analysis of HslV sequences, codon usage patterns, and genomic context across Chromobacterium species can reveal instances of horizontal gene transfer versus vertical inheritance, providing insights into the evolutionary history of this proteolytic system within the genus.

Methodological Approach for Evolutionary Analysis:

  • Construct phylogenetic trees based on:

    • HslV protein sequences

    • HslU protein sequences

    • Whole-genome sequences

  • Compare these trees to identify congruence or incongruence that might indicate different evolutionary pressures

  • Calculate selection pressures (dN/dS ratios) across the HslV sequence to identify regions under positive or purifying selection

  • Correlate sequence variations with ecological niches and pathogenic potential

This evolutionary perspective provides context for understanding how protein quality control systems contribute to bacterial adaptation and virulence across the Chromobacterium genus.

What are the implications of HslV research for understanding bacterial adaptation to environmental stresses?

Research on C. violaceum HslV has significant implications for understanding broader bacterial adaptation mechanisms to environmental stresses:

Stress Response Integration:
HslV represents a critical component of bacterial protein quality control networks that interface with multiple stress response pathways. Understanding how HslV activity is regulated during different stress conditions (heat shock, oxidative stress, nutrient limitation) provides insights into how bacteria integrate multiple stress signals to mount coordinated responses. The heat-shock regulation of HslV demonstrates how bacteria prioritize protein quality control during stress conditions.

Host-Pathogen Interface Adaptations:
For opportunistic pathogens like C. violaceum, HslV likely plays a crucial role in adaptation at the host-pathogen interface by:

  • Facilitating survival during transitions from environmental reservoirs to mammalian hosts

  • Managing proteomic damage caused by host immune defenses, particularly oxidative burst

  • Contributing to virulence through regulation of key pathogenicity factors, potentially including T3SS components

Ecological Niche Specialization:
The functional characteristics of HslV across different Chromobacterium species may reflect adaptations to their specific ecological niches:

  • Species associated with insect pathogenicity may show HslV adaptations for processing specific host proteins

  • Plant-associated species may have evolved specialized substrate specificities related to plant colonization

  • Species that cause opportunistic human infections like C. violaceum may possess HslV variants optimized for mammalian host temperatures and conditions

Climate Change Relevance:
Understanding how protein quality control systems like HslV enable bacterial adaptation has implications for predicting bacterial responses to climate change:

  • Temperature-dependent regulation of HslV provides insights into how bacteria might respond to changing global temperatures

  • HslV's role in managing damaged proteins informs predictions about bacterial adaptation to increased UV radiation and other environmental stressors

  • The connection between stress adaptation and virulence mediated by systems like HslV has implications for forecasting changes in bacterial pathogenicity under altered climate conditions

Methodological Implications:
The dual role of HslV in both basic cellular maintenance and virulence highlights the need for integrative experimental approaches that combine:

  • Laboratory stress simulations that mimic environmental conditions

  • In vivo infection models that capture host-specific stresses

  • Systems biology approaches that place HslV within global stress response networks

  • Ecological sampling to correlate HslV variants with environmental parameters

These insights from C. violaceum HslV research contribute to a broader understanding of bacterial stress adaptation mechanisms with implications for ecology, evolution, and infectious disease dynamics.

What are the future research directions for C. violaceum HslV studies?

Research on C. violaceum HslV is poised for significant advances in several key directions:

Structural Biology Frontiers:
Future research should focus on obtaining high-resolution structures of C. violaceum HslV-HslU complexes, particularly in substrate-bound states using cryo-electron microscopy. This structural information will enable precise mapping of substrate recognition sites and provide templates for rational drug design targeting this bacterial protease system.

Systems Biology Integration:
Developing comprehensive models of how HslV functions within C. violaceum's broader proteostasis network represents an important future direction. This includes mapping interactions between HslV and other protein quality control systems like chaperones, identifying regulatory networks controlling HslV expression, and understanding how these systems collectively respond to different environmental stresses.

Host-Pathogen Interaction Studies:
A critical area for future investigation is elucidating the precise role of HslV in C. violaceum pathogenesis. This includes determining whether HslV is required for:

  • Survival within specific host cell types

  • Resistance to particular host defense mechanisms

  • Regulation of established virulence factors like the T3SS

  • Processing of specific host proteins during infection

Therapeutic Applications:
The distinct characteristics of bacterial HslV compared to the eukaryotic proteasome make it a potential target for selective antimicrobial development. Future research should explore:

  • Development of specific inhibitors targeting bacterial HslV while sparing the eukaryotic proteasome

  • Assessment of HslV inhibition on C. violaceum virulence in animal models

  • Evaluation of potential synergies between HslV inhibitors and conventional antibiotics

Environmental Microbiology Applications:
Exploring how HslV contributes to C. violaceum's abundant presence in tropical and subtropical environments could provide insights into bacterial community dynamics and ecosystem functions. This includes investigating HslV's role in:

  • Interspecies competition within soil and aquatic microbial communities

  • Adaptation to fluctuating environmental conditions

  • Potential contributions to bioremediation capabilities

These future research directions will advance our fundamental understanding of bacterial proteolytic systems while potentially yielding new strategies for controlling C. violaceum infections and other bacterial pathogens.

How can understanding C. violaceum HslV contribute to broader fields in molecular microbiology?

Research on C. violaceum HslV has significant implications that extend beyond this specific system to impact broader fields in molecular microbiology:

Evolutionary Proteomics:
The HslV-HslU system represents an evolutionary intermediate between simpler bacterial proteases and the more complex eukaryotic proteasome. Detailed characterization of C. violaceum HslV provides a valuable reference point for understanding protease evolution across domains of life, offering insights into how complex proteolytic machines evolved from simpler ancestors.

Bacterial Stress Response Paradigms:
HslV research contributes to our understanding of how bacteria coordinate multiple protein quality control systems during stress adaptation. This generates broadly applicable principles regarding how cells prioritize protein degradation versus refolding under different stress conditions, how ATP-dependent proteolysis is regulated, and how these systems interface with transcriptional responses.

Virulence Regulation Mechanisms:
The potential connection between HslV and virulence factor expression in C. violaceum expands our understanding of post-translational regulation in bacterial pathogenesis. This contributes to emerging paradigms about how proteolytic systems can function as master regulators of virulence networks across diverse bacterial pathogens by controlling the stability of key transcriptional regulators and structural proteins.

Drug Development Approaches:
Insights from C. violaceum HslV structure-function relationships inform broader strategies for developing inhibitors targeting bacterial proteases. The principles derived from studying substrate recognition and catalytic mechanisms in this system can guide approaches for targeting other essential bacterial proteases as alternatives to conventional antibiotics in the face of rising antimicrobial resistance.

Environmental Adaptation Mechanisms:
Understanding how HslV contributes to C. violaceum's remarkable adaptability across different environmental niches enriches ecological models of bacterial persistence and community dynamics. This generates testable hypotheses about how proteolytic systems contribute to bacterial fitness in complex environments, with implications for predicting bacterial community responses to environmental changes.

By contributing to these broader fields, C. violaceum HslV research transcends its specific biological context to advance fundamental principles in molecular microbiology with far-reaching implications for bacterial physiology, evolution, ecology, and pathogenesis.

What specialized techniques are essential for comprehensive HslV functional characterization?

Comprehensive functional characterization of C. violaceum HslV requires integration of several specialized techniques:

Advanced Enzymatic Assays:

  • Real-time degradation assays using internally quenched fluorogenic peptides that allow continuous monitoring of proteolytic activity with high sensitivity

  • Single-molecule enzymology to characterize the processivity and dynamics of individual HslV-HslU complexes during substrate degradation

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map conformational changes in HslV upon ATP binding and substrate engagement

Structural Biology Approaches:

  • Cryo-electron microscopy with image classification to capture different conformational states of the HslV-HslU complex during the catalytic cycle

  • X-ray crystallography of HslV with bound inhibitors or substrate analogs to map the active site architecture

  • Hydrogen-deuterium exchange mass spectrometry to identify regions undergoing conformational changes during substrate processing

Substrate Identification Methods:

  • SILAC-based proteomics comparing protein turnover rates between wild-type and ΔhslV strains

  • Trap mutant approaches using catalytically inactive HslV variants to capture substrates

  • Ubiquitin-like protein tagging systems adapted for bacterial proteases to monitor substrate flux through the HslV-HslU system

In Vivo Activity Assessment:

  • Fluorescence-based protein degradation reporters expressed in C. violaceum to monitor HslV activity in living cells

  • Split fluorescent protein complementation to visualize HslV-substrate interactions in vivo

  • Selective protein degradation technologies using engineered HslV variants to target specific proteins and assess phenotypic consequences

Genetic Manipulation Approaches:

  • CRISPR-Cas9 genome editing for precise modification of hslV and associated genes

  • Inducible degradation systems to control HslV levels with temporal precision

  • Site-specific recombination systems for controlled expression of HslV variants in different genetic backgrounds

These specialized techniques, when integrated into a comprehensive research program, provide multidimensional insights into HslV function that go beyond traditional biochemical characterization, revealing dynamic aspects of proteolysis that are critical for understanding HslV's cellular roles.

What bioinformatic resources are available for analyzing HslV sequences and predicting substrate interactions?

Comprehensive Bioinformatic Resources for HslV Analysis:

Sequence Analysis Resources:

  • Specialized Protease Databases:

    • MEROPS (https://www.ebi.ac.uk/merops/) - Comprehensive protease classification system with HslV family information

    • ProteaseDB - Repository of protease sequences with functional annotations

    • ClanTox - Tool for identifying potential protease substrates based on sequence features

  • Evolutionary Analysis Tools:

    • MEGA X - Software package for comparative sequence analysis and phylogenetic tree construction

    • Phylogeny.fr - Automated phylogenetic analysis pipeline suitable for HslV evolutionary studies

    • ConSurf - Server for identifying conserved functional regions in HslV across species

Structure Prediction Resources:

  • AlphaFold2/RoseTTAFold - State-of-the-art protein structure prediction systems for modeling HslV structures from species lacking experimental structures

  • SWISS-MODEL - Homology modeling server useful for generating HslV structural models based on existing templates

  • 3DLigandSite - For predicting ligand binding sites in HslV models

  • DynaMut - For predicting effects of mutations on HslV protein stability and dynamics

Substrate Prediction Tools:

  • SitePrediction - For identifying potential cleavage sites in putative substrates

  • PROSPER/NetChop - Proteolytic site prediction servers that can be trained with HslV specificity data

  • STRING/IntAct - Protein-protein interaction databases to identify potential HslV interactors

  • DeepDegron - Machine learning approach to identify degradation signals in proteins

HslV-Specific Resources:

  • HslVDB - Custom database of HslV sequences with annotations of catalytic residues and substrate-binding sites

  • ProteasomeDB - Database of proteasome-like proteases including HslV family members

  • Bacterial Proteolysis Prediction Server (BPPS) - Specialized tool for predicting bacterial protease specificity

Integrated Analysis Platforms:

  • Galaxy - Web-based platform for accessible bioinformatic analysis of HslV sequences

  • Jalview - Interactive sequence alignment and analysis tool for visualizing HslV conservation

  • PyMOL/Chimera - Molecular visualization software with scripting capabilities for analyzing HslV structural features

Implementation Strategy:
For optimal HslV analysis, researchers should implement a multi-stage bioinformatic pipeline:

  • Initial sequence collection and basic alignment using BLASTP and ClustalO

  • Refinement with structure-guided alignment using PROMALS3D

  • Evolutionary analysis using MEGA X and ConSurf

  • Structural modeling with AlphaFold2 or homology modeling

  • Substrate prediction using specialized algorithms trained on existing HslV data

  • Integration of results using visualization tools like Jalview and PyMOL

This comprehensive bioinformatic approach enables researchers to generate testable hypotheses about HslV function, evolution, and substrate specificity that can guide experimental design.

What safety considerations should researchers address when working with recombinant C. violaceum systems?

Comprehensive Safety Guidelines for C. violaceum Research:

Biosafety Classification and Containment:
C. violaceum is typically handled at Biosafety Level 2 (BSL-2) due to its status as an opportunistic pathogen capable of causing severe, potentially fatal infections in humans . Any recombinant work involving C. violaceum requires institutional biosafety committee approval as it involves both recombinant DNA and infectious agents .

Laboratory Safety Protocols:

  • Personal Protective Equipment (PPE):

    • Laboratory coat dedicated to BSL-2 work

    • Disposable gloves changed frequently and whenever contaminated

    • Eye protection when handling liquid cultures

    • Closed-toe shoes mandatory in the laboratory

    • Consider face shield when handling large volumes

  • Engineering Controls:

    • All work with live C. violaceum cultures must be performed in a certified Class II biological safety cabinet

    • Centrifugation must use sealed rotors or safety cups

    • HEPA-filtered vacuum lines with disinfectant traps when aspirating cultures

    • Segregated incubation areas for C. violaceum cultures

  • Specific Hazards and Precautions:

    • Avoid generating aerosols when handling cultures

    • Use plasticware instead of glassware where possible to reduce sharps hazards

    • Implement rigorous decontamination protocols for all materials contacting C. violaceum

    • Maintain detailed inventory of all C. violaceum strains and derivatives

Recombinant DNA Considerations:
When creating recombinant C. violaceum strains, particularly those with modified HslV:

  • Risk Assessment Factors:

    • Evaluate whether genetic modifications could potentially increase virulence

    • Consider whether introduced genes could transfer to other organisms

    • Assess whether modified HslV might alter bacterial stress resistance or host interactions

  • Containment Strategies:

    • Use non-conjugative, non-mobilizable plasmid vectors when possible

    • Implement biological containment through auxotrophic or debilitated host strains

    • Consider inducible systems that prevent expression outside laboratory conditions

Emergency Response Procedures:

  • Spill Protocols:

    • Small spills: Cover with paper towels, apply appropriate disinfectant (e.g., 10% bleach), allow 20-minute contact time

    • Large spills: Evacuate area, notify laboratory supervisor, document incident

    • Personal exposure: Wash affected area with soap and water for 15 minutes, seek medical attention

  • Exposure Response:

    • Report any potential exposures immediately, even seemingly minor incidents

    • Document route of exposure and strain details for medical personnel

    • Provide medical professionals with information on C. violaceum susceptibility to antibiotics

Waste Management:

  • All material contacting C. violaceum must be decontaminated before disposal:

    • Liquid waste: Treat with 10% bleach (final concentration) for 30 minutes

    • Solid waste: Autoclave at 121°C for 45 minutes

    • Reusable items: Autoclave or disinfect with appropriate agent with verified efficacy

  • Maintain detailed waste disposal records in compliance with institutional requirements

Training Requirements:
Researchers working with C. violaceum must complete:

  • General biosafety training

  • Specific BSL-2 practices and procedures training

  • Laboratory-specific protocols for C. violaceum handling

  • Emergency response and spill management training

  • Documentation of training in laboratory records

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.