Protein L5 is indispensable for 50S subunit formation. Key findings include:
L5-deficient E. coli cells produce defective 45S ribosomal particles lacking 5S rRNA and CP-associated proteins (L16, L18, L25, etc.). These subunits cannot associate with 30S subunits to form functional ribosomes .
L5 facilitates 5S rRNA integration into the CP by forming cytoplasmic complexes with L18 and L25 prior to ribosomal incorporation .
Deletion of residues 73–80 in the P-site loop of L5 (uL5ΔPSL mutant) reduces growth rates by 40% and impairs cold adaptation in E. coli .
Mutant ribosomes exhibit:
| Parameter | Detail |
|---|---|
| Storage | -20°C (long-term: -80°C) |
| Stability | Avoid repeated freeze-thaw cycles |
| Species Reactivity | Enterococcus faecalis |
| Tag Information | N-/C-terminal tags (varies by lot) |
L5’s role in CP formation and tRNA positioning makes it a potential target for ribosomal inhibitors. Studies on recombinant L5 could inform drugs targeting:
This protein binds to and likely mediates the attachment of 5S rRNA to the large ribosomal subunit, forming part of the central protuberance. Within the 70S ribosome, it interacts with protein S13 of the 30S subunit (bridge B1b), connecting the two subunits. This bridge plays a role in subunit movement. It also interacts with the P-site tRNA; the 5S rRNA and associated proteins may contribute to stabilizing the positioning of ribosome-bound tRNAs.
KEGG: efa:EF0218
STRING: 226185.EF0218
The 50S ribosomal protein L5 (rplE) in Enterococcus faecalis is a critical component of the large ribosomal subunit that plays an essential role in ribosome assembly and function. It is specifically involved in the formation of the central protuberance (CP) of the large ribosomal subunit. Research has demonstrated that L5 is crucial for the proper assembly of the 50S subunit in bacterial cells . When L5 synthesis is arrested, bacterial cells develop defective large ribosomal subunits (45S particles) that lack most CP components, including 5S rRNA and several other proteins (L16, L18, L25, L27, L31, L33, and L35) . These defective subunits are unable to associate with the small ribosomal subunit, highlighting L5's importance in maintaining ribosomal structural integrity and function.
Expression of recombinant E. faecalis rplE protein typically involves several methodological approaches:
Bacterial Expression Systems:
E. coli-based expression: Most commonly used for initial protein expression studies due to high yield and established protocols.
E. faecalis expression: For more native conformation of the protein, expression in the original host may be preferred.
Enterococcus-specific expression systems:
E. faecalis MDXEF-1 can be modified as a host strain to express recombinant proteins .
The nisin-inducible expression system (NICE) can be utilized for controlled expression.
Expression verification methods:
Western blot analysis using anti-L5 antibodies
SDS-PAGE for protein size confirmation
Mass spectrometry for precise identification
The choice of expression system depends on research objectives, required protein folding, and downstream applications. For structural studies requiring native conformation, homologous expression in E. faecalis may provide advantages over heterologous systems.
Purification of recombinant E. faecalis rplE protein presents several methodological challenges:
Protein solubility issues:
L5 protein, as a ribosomal component, often forms inclusion bodies when overexpressed
Optimization of induction conditions (temperature, inducer concentration, induction time) is critical
Native binding partners:
L5 naturally binds to 5S rRNA and other ribosomal proteins, complicating purification
RNA contamination often occurs and requires specialized removal steps
Purification strategy:
Cell lysis (sonication or French press for bacterial cells)
Initial clarification (centrifugation to remove cell debris)
Affinity chromatography (His-tag, GST-tag, or other fusion tags)
Ion exchange chromatography (to separate based on charge properties)
Size exclusion chromatography (for final polishing and buffer exchange)
Quality control measures:
Protein purity assessment via SDS-PAGE (>95% purity typically required)
Functional assays to confirm biological activity
Circular dichroism to verify proper protein folding
Researchers often need to empirically determine optimal conditions for their specific construct and expression system to achieve sufficient yield and purity.
The absence of L5 in E. faecalis creates significant disruptions in ribosome assembly that follow both common bacterial patterns and species-specific alterations:
Common assembly defects across bacteria:
Formation of defective 45S particles instead of mature 50S subunits
Absence of central protuberance components (5S rRNA, L16, L18, L25, L27, L31, L33, and L35)
Inability of defective subunits to associate with the 30S subunit
E. faecalis-specific considerations:
Comparative genomic analysis suggests that E. faecalis may have unique ribosomal assembly factors not present in model organisms like E. coli
The central protuberance assembly in E. faecalis appears to follow the general bacterial model where L5 acts as a primary binding partner for 5S rRNA
Methodology for studying assembly differences:
Conditional knockdown of rplE gene in E. faecalis
Sucrose gradient centrifugation to isolate ribosomal particles
Quantitative mass spectrometry to determine protein composition of accumulated intermediates
Cryo-EM structural analysis of assembly intermediates
Comparative analysis with other bacterial species (e.g., E. coli, B. subtilis)
When L5 synthesis is arrested, 5S rRNA is found in the cytoplasm complexed with L18 and L25 proteins at quantities equal to the amount of ribosomes, suggesting that L5 is the limiting factor for incorporation of this entire subcomplex into the nascent 50S subunit . This assembly defect pattern appears to be conserved across bacterial species, though the precise kinetics and alternative assembly pathways may differ between E. faecalis and other bacteria.
Several sophisticated methodologies are employed to study the interactions between recombinant E. faecalis L5 protein and 5S rRNA:
In vitro binding assays:
| Method | Advantages | Limitations | Key Parameters |
|---|---|---|---|
| Electrophoretic Mobility Shift Assay (EMSA) | Simple, widely accessible | Semi-quantitative, not in solution | Protein:RNA ratio, buffer composition |
| Filter Binding Assay | Quantitative, rapid | May underestimate complex interactions | Kd determination, washing conditions |
| Surface Plasmon Resonance (SPR) | Real-time kinetics, label-free | Surface attachment may alter binding | Association/dissociation rates, binding affinity |
| Isothermal Titration Calorimetry (ITC) | Direct thermodynamic parameters | Requires large sample amounts | ΔH, ΔS, binding stoichiometry |
| Microscale Thermophoresis (MST) | Low sample consumption, solution-based | Requires fluorescent labeling | Temperature gradient, fluorescence detection |
Structural analysis techniques:
X-ray crystallography for atomic-level interaction details
Nuclear Magnetic Resonance (NMR) for solution structure and dynamics
Cryo-Electron Microscopy for visualization in near-native conditions
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for mapping interaction surfaces
Computational approaches:
Molecular dynamics simulations to predict binding energetics
RNA-protein docking algorithms to model complex formation
Sequence covariation analysis to identify co-evolving residues
When implementing these methods, researchers should consider using multiple complementary approaches to overcome the limitations of individual techniques. For example, combining EMSA for initial binding assessment with ITC for thermodynamic characterization and structural studies for mechanistic insights provides a comprehensive understanding of the L5-5S rRNA interaction.
Engineering recombinant E. faecalis to express L5 protein for vaccine delivery applications involves several methodological considerations:
Expression system design:
Surface display vs. secretion vs. intracellular expression strategies
Selection of appropriate promoters (constitutive or inducible)
Codon optimization for enhanced expression in E. faecalis
Fusion constructs (e.g., with dendritic cell targeting peptides) to enhance immunogenicity
Genetic engineering approaches:
Signal peptide addition for secretion pathways
Integration into the chromosome vs. plasmid-based expression
Use of food-grade selection markers for vaccine applications
Immunological considerations:
Fusion with dendritic cell targeting peptides significantly enhances immune responses
Recombinant E. faecalis expressing fusion proteins induces higher levels of specific antibodies (IgG and secretory IgA)
Higher proliferation of peripheral blood lymphocytes observed with properly designed constructs
Th1/Th2-type cytokine profiles can be modulated by expression system design
Safety assessment parameters:
Strain selection (community-associated clade B strains are preferable as they are commonly found in healthy individuals and rarely cause infections)
Genomic analysis to confirm absence of virulence factors and antibiotic resistance genes
Stability of the genetic construct in vivo
Colonization potential and persistence in the gastrointestinal tract
Research has demonstrated that E. faecalis can be effectively used as a delivery vehicle for antigens, with constructs carrying dendritic cell targeting peptides showing superior immune response induction compared to standard surface display . The E. faecalis strain MDXEF-1 has proven particularly effective as it can partially colonize the ceca, providing sustained stimulation of antigen-specific immune responses .
Studying the effects of L5 protein mutations on ribosome assembly in E. faecalis requires comprehensive methodological approaches:
Mutation strategies:
Site-directed mutagenesis of conserved residues
Domain swapping with L5 from other species
Deletion analysis of functional motifs
Random mutagenesis followed by functional screening
Expression systems for mutant analysis:
Complementation of L5-depleted strains
Conditional expression systems (temperature-sensitive, inducible)
Competitive expression with wild-type L5
Assembly analysis techniques:
| Technique | Application | Key Information Obtained | Limitations |
|---|---|---|---|
| Sucrose gradient centrifugation | Ribosomal profile analysis | Distribution of ribosomal particles (70S, 50S, 30S, assembly intermediates) | Limited resolution of intermediates |
| Quantitative mass spectrometry | Protein composition analysis | Precise stoichiometry of proteins in ribosomal particles | Sample preparation complexity |
| Cryo-EM | Structural analysis | 3D visualization of assembly defects | Resource intensive |
| In vivo ribosome assembly kinetics | Assembly timeline | Rate of formation of 50S subunits | Requires pulse-chase labeling |
| RNA-protein interaction assays | Binding affinity | Changes in L5-5S rRNA interaction | In vitro conditions may not reflect in vivo |
Phenotypic assessment:
Growth rate analysis under different conditions
Antibiotic sensitivity profiling (especially those targeting the ribosome)
Translation fidelity measurements using reporter systems
Stress response activation patterns
When L5 synthesis is arrested, the accumulation of defective 45S particles lacking central protuberance components provides a baseline for comparison . Mutations that affect L5-5S rRNA interactions would be expected to show similar defects to L5 depletion, with 5S rRNA and associated proteins (L18, L25) found in the cytoplasm rather than incorporated into the ribosome . Carefully designed mutation studies can provide insights into which domains of L5 are essential for specific steps in ribosome assembly.
The expression levels of recombinant L5 protein in E. faecalis vary significantly between different induction systems, with several factors affecting optimization:
Comparison of induction systems:
| Induction System | Advantages | Limitations | Typical Yield | Best Applications |
|---|---|---|---|---|
| Nisin-inducible (NICE) | Tight regulation, dose-dependent | Requires regulatory genes | High | Toxic protein expression |
| Constitutive (P23, P32) | Simple design, constant expression | No regulation | Moderate | Well-tolerated proteins |
| Xylose-inducible | Sugar-based induction (food-grade) | Lower dynamic range | Moderate | Food applications |
| Temperature-sensitive | No chemical inducer needed | Background expression | Variable | Large-scale production |
| pH-regulated | Environmentally responsive | Variable in complex media | Low-moderate | GI tract delivery |
Critical optimization factors:
Growth phase influence: Expression during exponential vs. stationary phase
Media composition: Rich vs. minimal media effects on protein yield
Temperature effects: Lower temperatures (25-30°C) often improve folding
Inducer concentration: Dose-response relationship for optimal expression
Expression duration: Time-course optimization to prevent degradation
Strain-specific considerations:
E. faecalis MDXEF-1 appears to produce nisin or nisin-like substances that can continuously induce expression of target proteins without external induction
Community-associated clade B strains may show different expression characteristics than hospital-associated clade A1 strains
Genome-sequenced strains allow for better prediction of potential interference from endogenous systems
Methodological approaches for optimization:
Factorial design experiments to simultaneously test multiple parameters
Reporter systems (GFP, luciferase) for real-time monitoring
Western blot quantification with internal standards
Flow cytometry for single-cell expression analysis
Resolving contradictory findings in E. faecalis L5 protein interaction studies requires systematic methodological approaches:
Sources of contradictory results:
Strain differences: Hospital-associated (clade A1), animal-associated (clade A2), and community-associated (clade B) strains may show different L5 interaction patterns
Experimental conditions: Buffer composition, salt concentration, and temperature significantly affect ribosomal protein interactions
Protein preparation methods: Different purification strategies may yield proteins with varying activity
In vitro vs. in vivo studies: Simplified in vitro systems may not capture the complexity of cellular environments
Systematic resolution approach:
| Step | Methodology | Expected Outcome |
|---|---|---|
| Standardize experimental conditions | Create detailed protocols with controlled parameters | Reduce variation between labs |
| Cross-validate with multiple techniques | Apply orthogonal methods (e.g., biochemical, structural, genetic) | Confirm findings through independent approaches |
| Perform strain comparisons | Test multiple E. faecalis isolates under identical conditions | Identify strain-specific differences |
| Meta-analysis | Systematically review published literature with statistical methods | Identify patterns in conflicting results |
| Collaborative replication | Independent verification by different laboratories | Establish reproducibility |
Data integration strategies:
Bayesian statistical approaches to weight evidence from multiple sources
Machine learning algorithms to identify patterns in complex datasets
Structural modeling to reconcile biochemical observations with physical constraints
By systematically addressing these factors, researchers can resolve apparent contradictions and develop a more cohesive understanding of E. faecalis L5 protein interactions within the ribosomal assembly pathway.
Assessing successful incorporation of recombinant L5 protein into E. faecalis ribosomes requires reliable biomarkers and analytical techniques:
Direct incorporation biomarkers:
| Biomarker | Methodology | Advantages | Limitations |
|---|---|---|---|
| Tagged L5 detection in purified ribosomes | Western blot, mass spectrometry | Direct evidence of incorporation | Tag may affect incorporation |
| L5:5S rRNA ratio | qRT-PCR, Northern blot | Quantitative assessment | Doesn't confirm functional assembly |
| Cryo-EM visualization | Structural imaging | Direct visualization of L5 position | Resource intensive, complex analysis |
| Ribosome profiling | Next-generation sequencing | Genome-wide translation analysis | Indirect measure of function |
| Crosslinking studies | Mass spectrometry analysis | Identifies interaction partners | Chemical modifications required |
Functional biomarkers:
Translation efficiency: Reporter systems (luciferase, GFP) to measure protein synthesis rates
Antibiotic sensitivity: Specific antibiotics targeting regions near L5 binding site
Growth kinetics: Restoration of normal growth in L5-depleted strains
Polysome formation: Polysome profiling to assess active translation complexes
Central protuberance assembly: Presence of complete CP components (L16, L18, L25, L27, L31, L33, and L35)
Integrated assessment approach:
Begin with sucrose gradient centrifugation to isolate 70S, 50S, and 30S particles
Analyze protein composition of 50S particles by quantitative mass spectrometry
Confirm L5 incorporation by Western blot with anti-L5 antibodies
Verify 5S rRNA incorporation by Northern blot
Assess functional translation capacity through in vitro translation assays
When L5 is successfully incorporated, the defective 45S particles should be replaced by proper 50S subunits capable of associating with 30S subunits to form 70S ribosomes . Additionally, 5S rRNA should shift from cytoplasmic complexes with L18 and L25 to ribosome incorporation . Monitoring these transitions provides reliable evidence of successful L5 incorporation into functional ribosomes.
Differentiating between pathogenic and probiotic potential in engineered E. faecalis strains requires comprehensive safety assessment methodologies:
Genomic analysis approaches:
Whole genome sequencing to identify:
Comparative genomics to distinguish:
Functional safety assessment:
| Assessment | Methodology | Key Indicators | Interpretation |
|---|---|---|---|
| Adhesion properties | Cell culture adhesion assays | Binding to intestinal cells | Moderate adhesion is beneficial; excessive may indicate pathogenic potential |
| Invasion capability | Gentamicin protection assay | Internalization into epithelial cells | Low/no invasion indicates safety |
| Cytotoxicity | MTT/LDH assays on cell lines | Cell death measurement | Minimal cytotoxicity required for safety |
| Inflammatory response | Cytokine profiling | Pro/anti-inflammatory balance | Anti-inflammatory profile preferred |
| Hemolytic activity | Blood agar plates | Hemolysis patterns | Absence of β-hemolysis preferred |
| Antibiotic susceptibility | MIC determination | Resistance profiles | Susceptibility to common antibiotics required |
Probiotic potential markers:
Resistance to gastrointestinal conditions (acid, bile)
Competitive exclusion of pathogens
Immunomodulatory properties
Absence of transmissible antibiotic resistance genes
Risk assessment framework:
Source strain evaluation (community-associated clade B strains are preferable as they rarely cause infections)
Genetic modification assessment (impact of L5 overexpression on virulence)
Comprehensive safety testing as outlined above
Animal model validation before human applications
Recent research has established methods to distinguish between hospital-associated clade A1 (rarely found in healthy individuals), animal-associated clade A2, and community-associated clade B (commonly found in healthy individuals and rarely causes infections) . Genomic comparison has demonstrated differential clustering of commensal and clinical isolates, suggesting these strains are specifically adapted to their respective environments . These advances in molecular biology allow for improved strain selection for recombinant protein expression applications.
Creating stable E. faecalis strains expressing recombinant L5 protein requires careful consideration of expression systems, genetic stability, and selection strategies:
Expression system selection:
| Expression System | Stability Features | Best Applications | Limitations |
|---|---|---|---|
| Chromosomal integration | Highest stability, defined copy number | Long-term studies, in vivo applications | Lower expression levels |
| Low-copy plasmids | Moderate stability with selection | Balance of expression and stability | Requires continuous selection |
| High-copy plasmids | Highest expression levels | Short-term, high-yield applications | Genetic instability, metabolic burden |
| Food-grade systems | No antibiotic resistance markers | Food and probiotic applications | Limited selection options |
Integration methodologies:
Homologous recombination:
Target neutral genomic locations (intergenic regions)
Use temperature-sensitive plasmids for selection/counterselection
Two-step process with verification by PCR and sequencing
CRISPR-Cas9 assisted integration:
Higher efficiency targeted integration
Reduced off-target effects
Marker-free integration possible
Expression stabilization strategies:
Codon optimization for E. faecalis (especially important for L5 protein)
Balanced promoter strength to minimize metabolic burden
Use of terminators to prevent read-through transcription
Strategic placement of regulatory elements
Selection and maintenance protocols:
Initial selection with appropriate antibiotics for transformant isolation
Passage without selection to test stability (minimum 100 generations)
Quantification of retention rate by selective plating
Regular verification of expression levels after extended culture
Quality control procedure:
Colony PCR screening for correct integrants
Whole genome sequencing to confirm single-site integration
RT-qPCR to verify stable transcript levels
Western blot analysis for consistent protein expression
Functional testing of L5 incorporation into ribosomes
For maximum stability in E. faecalis strains intended for long-term research applications, chromosomal integration using CRISPR-Cas9 assisted homologous recombination into a neutral site, with a moderately strong constitutive promoter, has proven most effective. When utilizing E. faecalis MDXEF-1, researchers should consider its potential self-induction capabilities which may affect expression levels over time .
Accurately measuring the impact of recombinant L5 protein expression on E. faecalis ribosome function and protein synthesis requires sophisticated methodological approaches:
Translation efficiency assessment:
| Method | Application | Advantages | Considerations |
|---|---|---|---|
| Ribosome profiling | Genome-wide translation analysis | Nucleotide resolution, quantitative | Complex bioinformatics analysis |
| Polysome profiling | Global translation state | Established methodology | Limited resolution |
| Reporter systems | Targeted measurement | Simple implementation | Limited to specific mRNAs |
| Puromycin incorporation | Global protein synthesis rate | Quick assessment | Lacks mRNA specificity |
| 35S-methionine labeling | De novo protein synthesis | Direct quantification | Radioactivity, limited resolution |
Ribosome functionality assays:
In vitro translation systems:
Reconstituted translation using purified components
Measurement of specific protein synthesis rates
Assessment of translation accuracy (stop codon readthrough, frameshifting)
Ribosome structural integrity:
Kinetic measurements:
Translation elongation rate determination using ribosome profiling
Initiation efficiency assessment with toeprinting assays
Termination accuracy evaluation with dual-luciferase reporters
Physiological impact assessment:
Growth rate analysis under various stress conditions
Competition assays with wild-type strains
Antibiotic sensitivity testing (especially translation-targeting antibiotics)
Proteome analysis by mass spectrometry to detect global changes
Experimental design considerations:
Use inducible expression systems to create dose-response relationships
Include appropriate controls (empty vector, inactive L5 mutants)
Account for potential autoregulation of endogenous L5 expression
Consider the temporal dynamics of ribosome assembly and turnover
Given that L5 is essential for proper formation of the central protuberance of the 50S subunit , excess or mutant L5 expression could potentially interfere with ribosome assembly. By applying these methodologies, researchers can quantitatively determine whether recombinant L5 expression enhances ribosome function, creates dysfunctional ribosomes, or alters the balance of mature vs. immature ribosomal particles.
The field of recombinant E. faecalis L5 protein research presents several promising future research directions:
Structural biology and ribosome assembly:
Cryo-EM studies of assembly intermediates to visualize the precise role of L5 in central protuberance formation
Single-molecule studies of L5-5S rRNA interactions during ribosome assembly
Comparative structural analysis of L5 proteins across bacterial clades to understand evolutionary adaptations
Therapeutic applications:
Development of L5-targeted antimicrobials that specifically disrupt ribosome assembly
Exploration of L5 as a carrier protein for antigenic epitopes in vaccine development
Investigation of L5-based inhibitors of protein synthesis in pathogenic bacteria
Biotechnology applications:
Engineering ribosomes with modified L5 proteins for expanded genetic code applications
Development of biosensors based on L5-dependent ribosome assembly
Creation of L5 variants with enhanced protein synthesis capabilities for biotechnology applications
Vaccine delivery systems:
Further optimization of E. faecalis as a mucosal vaccine delivery vehicle using L5 fusion proteins
Development of multi-antigen presentation systems using the L5 scaffold
Exploration of dendritic cell targeting peptide fusions with L5 to enhance immune responses
Safety and regulatory considerations:
Comprehensive genomic analysis to distinguish between safe and potentially harmful E. faecalis strains
Development of biocontainment strategies for engineered E. faecalis strains
Establishment of standard protocols for safety assessment of recombinant E. faecalis strains
Research has demonstrated that E. faecalis can effectively deliver antigens and stimulate robust immune responses, particularly when constructed with dendritic cell targeting peptides . Further exploration of L5 as a component in these delivery systems, especially in combination with other ribosomal proteins or bacterial antigens, holds significant promise. Additionally, the critical role of L5 in ribosome assembly makes it an attractive target for antimicrobial development, where inhibition of proper L5 function could prevent formation of functional ribosomes in pathogenic bacteria.
Addressing biosafety concerns with recombinant E. faecalis strains requires comprehensive risk assessment and management strategies:
Strain selection and engineering considerations:
Biosafety assessment protocols:
Genomic characterization:
Comprehensive screening for virulence factors
Antibiotic resistance gene profiling
Mobile genetic element identification
Phenotypic safety testing:
Hemolysis assays
Cytotoxicity testing on mammalian cell lines
Invasion capacity in epithelial cell models
Inflammatory response assessment (cytokine profiles)
Containment verification:
Environmental survival studies
Horizontal gene transfer frequency measurement
Stability assessment under non-selective conditions
Regulatory compliance strategies:
Development of well-characterized, safety-enhanced E. faecalis strains
Implementation of biological containment systems
Creation of strain-specific detection methods for environmental monitoring
Detailed documentation of genetic modifications and safety testing results
Ethical considerations:
Transparent risk communication with relevant stakeholders
Appropriate biosafety level classification for research activities
Responsible innovation framework for technology development
Consideration of dual-use potential
Recent advances in genomic analysis have enabled the distinction between hospital-associated clade A1, animal-associated clade A2, and community-associated clade B strains of E. faecalis . This classification provides a scientific basis for strain selection, with community-associated clade B strains representing the safest option for biotechnology applications. Additionally, comparative genomic studies have demonstrated differential clustering of commensal and clinical isolates , providing further guidance for identifying strains with minimal pathogenic potential.