Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Tag type is determined during production. Please specify your preferred tag type for prioritized development.
KEGG: efa:EF1647
STRING: 226185.EF1647
HslV functions as the proteolytic component of the HslU-HslV protease system in Enterococcus faecalis. It forms a complex with the ATP-dependent HslU subunit to create a functional proteolytic machinery that plays a critical role in protein quality control and degradation of misfolded proteins. The HslV component specifically serves as the peptidase proteolytic subunit within this system and is classified as part of the broader ATP-dependent protease family in E. faecalis . This protease system bears structural and functional similarities to the eukaryotic proteasome, making it an important subject for comparative studies of protein degradation mechanisms across different domains of life.
The HslV-HslU complex functions through a coordinated mechanism where HslU (the ATPase subunit) recognizes, unfolds, and translocates substrate proteins into the proteolytic chamber formed by HslV. The process begins with substrate recognition by HslU, followed by ATP-dependent conformational changes that facilitate protein unfolding. The unfolded polypeptide is then threaded into the central chamber of the HslV component, where it undergoes proteolytic cleavage. This ATP-driven proteolysis system represents an essential quality control mechanism in E. faecalis, contributing to the degradation of misfolded, damaged, or regulatory proteins . The functional interaction between these subunits is critical for the proper maintenance of protein homeostasis within the bacterial cell.
For optimal cloning of E. faecalis hslV, begin by designing primers that include appropriate restriction sites compatible with your expression vector. Consider adding a C-terminal or N-terminal affinity tag (His-tag is commonly used) to facilitate purification, though care should be taken to ensure the tag doesn't interfere with protein folding or activity. For E. faecalis genes, codon optimization for your expression host may be necessary, especially when expressing in E. coli, since there are differences in codon usage between these organisms. Similar to approaches used for other E. faecalis proteins, you can employ PCR amplification using high-fidelity DNA polymerase and genomic DNA as template . After restriction digestion and ligation into your chosen expression vector, transformation procedures similar to those described for E. coli can be applied using established techniques . Verify your construct through restriction analysis and sequencing before proceeding to expression trials.
Codon optimization can significantly improve the expression of E. faecalis hslV in heterologous systems by addressing several critical factors. E. faecalis has a different GC content and codon usage bias compared to common expression hosts like E. coli, which can lead to translational pausing, premature termination, or reduced protein yields when expressing the native sequence. Optimizing the codons to match the preferred codons of your expression host can enhance translation efficiency and increase protein yield by 5-10 fold in many cases. When planning codon optimization for hslV, analyze the rare codons in the native sequence and modify them while maintaining the same amino acid sequence. Additionally, eliminate potential internal Shine-Dalgarno sequences, RNA secondary structures in the 5' region, and negative cis-regulatory elements that might impede translation. Similar methodological approaches to those used for expressing other E. faecalis genes should be applied , with careful validation of the optimized construct through small-scale expression trials before proceeding to larger-scale production.
For high-purity recombinant HslV from E. faecalis, a multi-step purification approach is recommended. Begin with affinity chromatography using a His-tagged construct and Ni-NTA resin, which provides good initial capture of the target protein. Use a lysis buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10 mM imidazole, and 1 mM DTT, with elution performed using an imidazole gradient (50-250 mM). Follow this with ion-exchange chromatography (typically Q-Sepharose for HslV) to separate the target protein from contaminants with different charge properties. Finally, apply size-exclusion chromatography (Superdex 200) to obtain highly pure protein and to confirm the oligomeric state of HslV, which typically forms a dodecameric structure. Throughout purification, use SDS-PAGE analysis to monitor protein purity and Western blotting to confirm identity. Similar to isolation techniques used for other E. faecalis components, it's important to include protease inhibitors in all buffers to prevent degradation during purification . For functional studies, co-purification or separate purification and reconstitution with the HslU subunit will be necessary as the complete HslU-HslV complex is required for full proteolytic activity.
For characterizing the oligomeric state of HslV and its complex with HslU from E. faecalis, analytical size-exclusion chromatography (SEC) serves as a primary method to determine the apparent molecular weight and complex formation. The HslV subunit typically forms a dodecamer, while HslU forms a hexamer, combining in a ratio of 1:1 to form the complete HslU-HslV complex. Native polyacrylamide gel electrophoresis (Native-PAGE) provides complementary information about the intact complex under non-denaturing conditions. Analytical ultracentrifugation (AUC), particularly sedimentation velocity experiments, offers precise determination of molecular weight and shape parameters of both individual components and the assembled complex. Multi-angle light scattering (MALS) coupled with SEC provides absolute molecular weight determination independent of shape assumptions. For visualizing the complex architecture, negative-stain electron microscopy or, for higher resolution, cryo-electron microscopy can be employed. Cross-linking mass spectrometry (XL-MS) can identify specific interaction interfaces between HslU and HslV subunits. These approaches should be coupled with functional assays to correlate oligomeric states with proteolytic activity, following methodological considerations similar to those applied for other bacterial ATP-dependent proteases.
For reliable assessment of recombinant E. faecalis HslV activity, several complementary assays should be employed. The primary approach involves using fluorogenic peptide substrates such as Suc-LLVY-AMC (succinyl-Leu-Leu-Val-Tyr-7-amido-4-methylcoumarin), which releases quantifiable fluorescence upon cleavage. This assay must be performed with reconstituted HslU-HslV complex, as HslV alone shows minimal activity. Set up reaction mixtures containing 0.1-1 μM HslV, 0.1-1 μM HslU, 100 μM fluorogenic substrate, 5 mM ATP, 15 mM MgCl₂, and 50 mM Tris-HCl (pH 8.0) at 37°C, with fluorescence measured over time (excitation 380 nm, emission 460 nm). Additionally, degradation of model protein substrates like casein-FITC or unfolded GFP can be monitored by SDS-PAGE or fluorescence decrease. ATP hydrolysis coupled with the proteolytic activity can be measured using a malachite green assay to detect released phosphate. For all assays, appropriate controls should include reactions without ATP, without HslU, and with known protease inhibitors to verify specificity. These methodological approaches are similar to those used for characterizing other ATP-dependent proteases and should be optimized specifically for the E. faecalis enzyme system.
ATP concentration exerts a profound influence on HslV-HslU proteolytic activity through multiple mechanisms. The system shows a sigmoidal relationship between ATP concentration and proteolytic activity, with typical optimal ATP concentrations ranging between 2-5 mM. At low ATP concentrations (<0.5 mM), the complex shows minimal activity as ATP binding is required for HslU conformational changes that enable substrate processing and activation of HslV. As ATP concentration increases (0.5-2 mM), activity rises sharply due to enhanced complex formation and substrate processing. At optimal concentrations (2-5 mM), maximum proteolytic rates are achieved as all available HslU subunits can bind ATP and undergo necessary conformational changes. At very high ATP concentrations (>10 mM), some inhibition may occur due to competition effects or alterations in complex stability. When designing experiments to characterize this relationship, use a buffer system containing 50 mM Tris-HCl (pH 8.0), 15 mM MgCl₂ (essential cofactor for ATP hydrolysis), and ATP concentrations ranging from 0-10 mM, testing proteolytic activity using standard fluorogenic peptide substrates. The detailed ATP concentration dependence should be determined through Michaelis-Menten kinetic analysis to obtain KM values for ATP and correlate them with structural changes in the complex.
The substrate specificity profile of E. faecalis HslV, when operating as part of the HslU-HslV complex, shows distinct characteristics compared to other bacterial proteases. This ATP-dependent protease demonstrates preference for hydrophobic and basic residues at the P1 position (the amino acid immediately preceding the cleavage site), with particular affinity for leucine, phenylalanine, tyrosine, and arginine residues. Unlike some other bacterial proteases such as Lon or ClpP, the HslV complex shows more restricted specificity, typically requiring longer peptide sequences for recognition. Compared to the well-studied E. coli HslV-HslU system, the E. faecalis homolog may exhibit subtle differences in specificity that reflect adaptations to its ecological niche. For comprehensive specificity profiling, employ peptide libraries with systematic amino acid substitutions at the P4-P4' positions surrounding the cleavage site. Analyze cleavage products using mass spectrometry to generate a position-specific scoring matrix. Additionally, compare degradation rates of model proteins with known structural properties to assess how secondary structure elements affect substrate processing. These methodological approaches should be adapted from general protease characterization techniques and tailored specifically to the ATP-dependent nature of the HslV-HslU system, ensuring that all assays include appropriate ATP regeneration systems.
Recombinant HslV provides a valuable tool for investigating antibiotic resistance mechanisms in E. faecalis through several research approaches. Proteases like HslV-HslU play crucial roles in stress response and protein quality control, which can indirectly impact antibiotic resistance. To leverage this for research, compare HslV-HslU activity in antibiotic-resistant strains versus susceptible strains, particularly focusing on linezolid-resistant isolates where mutations in ribosomal components have been identified . Design experiments to determine if the HslV-HslU system participates in degrading misfolded proteins that arise due to ribosomal mutations, potentially contributing to resistance phenotypes. Implement proteomics approaches to identify differential substrates of HslV-HslU in resistant versus susceptible strains, which might reveal novel resistance-associated proteins. Consider testing whether overexpression or deletion of hslV affects minimum inhibitory concentrations (MICs) of various antibiotics, particularly those targeting protein synthesis like linezolid. Additionally, investigate whether HslV-HslU activity changes during antibiotic exposure and stress conditions, and determine if the system influences homologous recombination rates, which are known to be important in the development of high-level linezolid resistance in E. faecalis as demonstrated by studies comparing recombination-proficient and recombination-deficient strains .
The HslV-HslU protease system likely plays multifaceted roles in E. faecalis virulence and biofilm formation through its function in protein quality control and stress response regulation. To investigate these connections, researchers should create hslV and hslU deletion mutants in clinically relevant E. faecalis strains and assess changes in biofilm formation capacity using crystal violet staining assays and confocal microscopy analysis. Compare the proteomes of wild-type and mutant strains under biofilm-inducing conditions to identify differentially expressed virulence factors that might be regulated by the HslV-HslU system. Examine the system's role in stress tolerance (oxidative, acid, antimicrobial peptides) that is relevant to survival in host environments using growth kinetics and viability assays under various stressors. Conduct in vitro infection models using epithelial cell lines or macrophages to assess changes in adhesion, invasion, and intracellular survival of hslV/hslU mutants. The HslV-HslU system may also influence genetic exchange within biofilms by affecting rates of homologous recombination, similar to observations in linezolid resistance development where recombination proficiency influences mutation frequencies . For in vivo relevance, utilize Caenorhabditis elegans or mouse models of E. faecalis infection to compare the virulence of wild-type and protease-deficient strains, measuring colonization, dissemination, and host survival rates.
Structural knowledge of E. faecalis HslV can be leveraged to develop innovative antimicrobial strategies targeting this essential protein quality control system. Begin by performing detailed structural characterization through X-ray crystallography or cryo-electron microscopy of the HslV-HslU complex, focusing on active site architecture and substrate binding pockets. Use this structural information to conduct in silico screening of chemical libraries to identify potential inhibitors that specifically target E. faecalis HslV, with particular attention to compounds that can distinguish between bacterial HslV and the human proteasome to minimize off-target effects. Validate candidate inhibitors through biochemical assays measuring HslV-HslU proteolytic activity using fluorogenic substrates, determining IC50 values and inhibition mechanisms (competitive, non-competitive, or allosteric). Examine structure-activity relationships through systematic modification of lead compounds to improve potency and selectivity. Evaluate cellular efficacy by testing inhibitor effects on E. faecalis growth, particularly under stress conditions where proteolysis becomes essential. For promising compounds, assess their ability to potentiate existing antibiotics, especially against resistant strains where mutations in ribosomal components like those observed in linezolid resistance might increase cellular dependence on protein quality control systems. This approach builds on successful strategies used for developing proteasome inhibitors in cancer therapy, adapted to the bacterial context.
Low solubility of recombinant E. faecalis HslV can be addressed through multiple strategies targeting expression conditions, buffer optimization, and protein engineering. For expression conditions, reduce induction temperature to 16-20°C and lower IPTG concentration to 0.1-0.2 mM to slow protein production and allow proper folding. Consider using specialized E. coli strains such as Arctic Express or Rosetta-gami that promote proper folding of challenging proteins. For buffer optimization, screen a range of pH conditions (pH 6.5-8.5) as HslV solubility can be highly pH-dependent, and test various salt concentrations (100-500 mM NaCl) to identify optimal ionic strength. Include solubility enhancers such as 5-10% glycerol, 0.5-1 M urea (non-denaturing concentration), or 50-100 mM arginine in your lysis and storage buffers. If these approaches are insufficient, consider protein engineering solutions such as expressing HslV with solubility-enhancing fusion partners like MBP (maltose-binding protein) or SUMO, with appropriate protease cleavage sites for tag removal after purification. Co-expression with its partner HslU may also enhance solubility by promoting proper complex formation. For extraction from inclusion bodies, develop a refolding protocol using step-wise dialysis from denaturing conditions (6 M urea) with gradually decreasing denaturant concentration, though this approach may result in lower recovery of active protein.
When troubleshooting issues with proteolytic activity of recombinant E. faecalis HslV-HslU, follow a systematic approach addressing complex formation, assay conditions, and protein integrity. First, verify that both HslV and HslU are present in the correct stoichiometric ratio (typically 1:1 complex of HslV dodecamer to HslU hexamer) using size-exclusion chromatography or native gel electrophoresis. Ensure ATP is fresh and present at optimal concentration (2-5 mM) with the required Mg²⁺ cofactor (10-15 mM). Optimize buffer conditions by testing different pH values (pH 7.0-8.5) and salt concentrations (50-200 mM), as ionic strength can significantly affect complex assembly and activity. Verify substrate quality and concentration, using established fluorogenic peptides like Suc-LLVY-AMC at 50-100 μM. For persistent activity issues, check for inhibitory contaminants in your protein preparation using mass spectrometry analysis, and consider additional purification steps if necessary. Examine protein integrity through SDS-PAGE and mass spectrometry to identify any degradation or post-translational modifications that might affect activity. If co-expressing both subunits, verify that both genes are being expressed properly through Western blotting. For very low activity, consider concentrating the complex using ultrafiltration devices (30-100 kDa cutoff) and adding ATP regeneration systems (creatine phosphate/creatine kinase) to maintain ATP levels during extended assays.
Batch-to-batch variability in recombinant E. faecalis HslV preparation can stem from multiple factors that should be systematically controlled. Expression conditions represent a primary source of variability; fluctuations in induction timing, cell density at induction, and harvest time can significantly affect protein quality. Standardize these parameters precisely, maintaining OD₆₀₀ at induction between 0.6-0.8 and fixed induction periods (e.g., exactly 16 hours at 18°C). Media composition differences between batches can alter expression levels and protein folding; use high-quality media components and prepare fresh media for each expression run. During purification, variations in buffer preparation, column performance, and fraction collection criteria can introduce inconsistencies. Implement detailed SOPs for buffer preparation with pH verification, regular column performance testing, and consistent fraction pooling criteria based on both chromatographic profiles and activity assays. Post-purification handling, including freeze-thaw cycles, storage temperature fluctuations, and protein concentration processes, can cause activity loss and aggregation. Aliquot purified protein to avoid repeated freeze-thaw cycles, maintain consistent storage conditions, and standardize concentration procedures. Implement quality control measures for each batch, including specific activity determination using standardized substrates, SDS-PAGE analysis for purity assessment, and dynamic light scattering to evaluate aggregation state. These methodological considerations should be documented in detailed batch records that allow traceability of any variations observed between preparations.
For comprehensive analysis of E. faecalis HslV substrate specificity and creation of detailed specificity profiles, implement a multi-faceted approach combining positional scanning libraries, proteomic methods, and computational analysis. Begin with positional scanning peptide libraries where one position is systematically varied while others remain constant, using fluorogenic or chromogenic reporters to quantify cleavage efficiency. This provides position-specific preferences at the P4 to P4' positions surrounding the scissile bond. For broader substrate identification, employ proteomic approaches such as PICS (Proteomic Identification of protease Cleavage Sites) where a complex protein mixture is digested by the HslV-HslU complex, and resulting peptides are identified by mass spectrometry. Terminal amine isotopic labeling of substrates (TAILS) can also be used to identify protein N-termini generated by HslV-HslU cleavage in complex mixtures. To create a comprehensive specificity profile, analyze the enrichment of particular amino acids at each position relative to their natural abundance, and generate sequence logos to visualize preferences. Validate results using designed synthetic peptides with systematic variations around identified cleavage sites. For in vivo relevance, compare your specificity data with the E. faecalis proteome to identify potential natural substrates, and validate these predictions through targeted degradation assays. These approaches should be coupled with structural analysis of substrate binding sites to rationalize observed specificities and potentially predict specificity differences between HslV from different bacterial species.
Proteomics data can be leveraged through several complementary approaches to identify natural substrates of the HslV-HslU system in E. faecalis. Implement a comparative proteomics strategy using SILAC (Stable Isotope Labeling with Amino acids in Cell culture) or TMT (Tandem Mass Tag) labeling to compare protein abundance in wild-type E. faecalis versus ΔhslV or ΔhslU mutants. Proteins that accumulate in the mutants represent potential natural substrates of the proteolytic system. To capture direct interactions, employ a substrate-trapping approach using catalytically inactive HslV (create by site-directed mutagenesis of active site residues) combined with crosslinking and immunoprecipitation followed by mass spectrometry (CLIP-MS). This identifies proteins that are recognized and bound but not degraded by the complex. For degradation kinetics, perform pulse-chase experiments with global proteome analysis to identify proteins with differential half-lives between wild-type and protease-deficient strains. Apply N-terminomics approaches such as TAILS (Terminal Amine Isotopic Labeling of Substrates) to identify specific cleavage sites generated by HslV-HslU activity in vivo. After identifying candidate substrates, validate them through in vitro degradation assays using purified recombinant proteins and reconstituted HslV-HslU complex. Analyze the validated substrates for common features including sequence motifs, structural elements, or cellular functions to develop a comprehensive understanding of HslV-HslU substrate recognition principles. Examine the proteomics data from E. faecalis in different growth conditions and stress responses to determine context-specific substrates, as the ATP-dependent proteases often display condition-dependent activity profiles.