Lactobacillus plantarum is a widespread probiotic bacterium commonly found in many fermented food products . It is known for producing diverse metabolites that play key roles in enhancing health and food quality, including peptides, organic acids, exopolysaccharides, and antimicrobial compounds, which contribute to gut health, immune system modulation, and pathogen inhibition .
Recombinant Lactobacillus plantarum is a genetically engineered strain of L. plantarum that has been modified to express specific genes or proteins . This technology has been employed to enhance the probiotic properties of L. plantarum or to use it as a delivery vehicle for therapeutic proteins .
Surface Display of Proteins: Recombinant L. plantarum can be engineered to display proteins on its surface, which can enhance its interaction with the host immune system or deliver therapeutic proteins directly to the target site .
Immune Modulation: Recombinant L. plantarum can be used to modulate the host immune response by expressing cytokines such as IL-10 .
Vaccine Delivery: Recombinant L. plantarum can be used as a vaccine delivery vehicle by expressing antigens from pathogens .
Lactobacillus plantarum has been shown to enhance IL-10 production . IL-10 is an anti-inflammatory cytokine that is vital in protecting the host from tissue damage during acute phases of the immune response toward a pathogenic infection . IL-10 exerts immunosuppression through antigen-presenting cells (APCs) and can significantly inhibit APCs, especially macrophages and dendritic cells (DCs) .
Lactobacillus plantarum PL-02 supplementation can improve muscle mass and exercise performance and reduce exercise fatigue . Studies have shown that PL-02 supplementation significantly decreases lactate, NH3, BUN, and CK levels .
A reference proteomic database of L. plantarum has been established using two-dimensional electrophoresis and tandem mass spectrometry . This database includes information on whole-cell proteins and secretory proteins, providing valuable information for further proteomic researches of L. plantarum .
Recombinant UPF0210 protein lp_2507 can be expressed and purified from multiple host systems, each offering distinct advantages:
E. coli expression systems: Provide high yields and shorter turnaround times, ideal for initial structural studies .
Yeast expression systems: Offer good yields with some post-translational modifications .
Baculovirus expression systems: Provide more complex eukaryotic post-translational modifications necessary for correct protein folding .
Mammalian cell expression systems: Offer the most extensive post-translational modifications, potentially retaining higher protein activity .
The selection of an appropriate expression system should be based on your specific research requirements, including needed protein yield, folding requirements, and post-translational modifications essential for function.
The purification strategy for recombinant lp_2507 typically involves:
Cell lysis: Using mechanical disruption methods like homogenization, particularly effective for bacterial cell walls .
Initial separation: Cell wall debris containing the protein can be isolated through centrifugation.
Heat treatment: The sample can be subjected to 100°C for 5 minutes when using Western blot analysis .
Affinity chromatography: If expressed with an affinity tag (commonly His-tag), this allows for selective purification.
Size exclusion chromatography: For further purification based on molecular size.
For surface-displayed proteins, specialized extraction techniques may be necessary to efficiently isolate the protein from the cell wall components.
Multiple complementary detection methods can be employed:
Western blot analysis: The primary method to confirm protein expression and determine molecular weight. Anti-His tag antibodies are commonly used if the protein contains a His-tag .
Fluorescence microscopy: If the protein is fused with GFP or FITC, direct visualization of expression and localization is possible .
Flow cytometry: Can quantify expression efficiency in a population of recombinant bacteria. Studies with similar recombinant proteins have shown expression rates of 37.5% in modified strains compared to 2.5% in parental strains .
Transmission electron microscopy (TEM): Useful for visualizing surface-displayed proteins and assessing whether protein expression affects bacterial morphology .
Optimization requires systematic evaluation of multiple parameters:
| Parameter | Optimization Range | Measurement Method |
|---|---|---|
| Induction time | 2-22 hours | Western blot quantification |
| Inducer concentration | 10-150 ng/mL | Protein yield assessment |
| Temperature | 30-37°C | Expression level and solubility |
| Bacterial passage | Passages 1-10 | Consistency of expression |
Studies with similar recombinant proteins in L. plantarum have shown that:
Highest protein yields were obtained with 50 ng/mL inducer concentration
Early passages (1-5) maintained consistent expression levels, while later passages showed variable results
When optimizing, it's crucial to assess not only quantity but also quality (folding, activity) of the expressed protein through functional assays specific to lp_2507.
Recombinant protein production imposes metabolic stress on host organisms at multiple levels:
Transcriptional burden: Even transcription without translation creates significant metabolic burden . Consider using:
Translational burden: Can contribute to metabolic stress, especially when:
Post-translational processing: Surface display systems like pgsA may increase burden
Mitigation strategies:
Use truncated anchor proteins (e.g., pgsA' instead of full-length pgsA) which has shown superior expression efficiency
Implement fed-batch cultivation to maintain optimal nutrient availability
Consider inducible systems that allow biomass accumulation before protein expression
Monitor growth curves to identify optimal harvest points before growth inhibition becomes severe
Recombinant proteins may encounter folding issues that affect functionality:
Computational analysis and rational design:
Analyze the lp_2507 sequence for potential folding obstacles
Identify domains that might benefit from flexible linkers
Remove or modify hydrophobic regions prone to aggregation
Co-expression strategies:
Express with molecular chaperones to assist folding
Consider fusion partners known to enhance solubility (e.g., thioredoxin, SUMO)
Expression conditions modifications:
Lower expression temperature (30°C instead of 37°C) to slow folding
Adjust media composition to provide necessary co-factors
Optimize inducer concentration to prevent overwhelming cellular machinery
Post-expression processing:
In vitro refolding protocols if the protein forms inclusion bodies
On-column refolding during purification
Success indicators should include not only solubility but functional assays to confirm proper folding has occurred.
To assess immunomodulatory properties of lp_2507, similar to studies with other L. plantarum surface proteins:
Co-culture experiments:
Prepare RAW264.7 macrophage cells or primary immune cells
Co-culture with recombinant L. plantarum expressing lp_2507
Include appropriate controls: non-recombinant L. plantarum, empty vector controls
Cytokine profiling:
Flow cytometry analysis:
Stimulation experiments:
These methodologies can determine whether lp_2507 has immunostimulatory or immunoregulatory properties and characterize its mechanism of action.
A comprehensive stability assessment should include:
Temperature stability testing:
Expose purified protein to temperature range (4°C, 25°C, 37°C, 50°C)
Test at multiple timepoints (24h, 48h, 72h, 1 week)
Assess remaining activity through functional assays
Analyze structural integrity via circular dichroism or thermal shift assays
pH stability assessment:
Test stability at pH range (1.5-8.0)
Important for applications involving gastrointestinal conditions
Quantify activity retention at each pH point
Salt concentration effects:
Evaluate stability in varying ionic strengths
Assess protein aggregation tendencies
Freeze-thaw stability:
Subject protein to multiple freeze-thaw cycles
Quantify activity loss per cycle
Determine optimal cryoprotectant formulations
Long-term storage optimization:
Compare different storage buffers
Test additives (glycerol, trehalose, albumin)
Establish shelf-life under optimized conditions
Results should be tabulated with specific activity retention percentages under each condition to develop evidence-based storage recommendations.
As an uncharacterized protein family member, systematic approaches are needed:
Bioinformatic analysis:
Perform sequence alignments with characterized proteins
Identify conserved domains that suggest function
Use structure prediction algorithms (AlphaFold2) to model 3D structure
Apply molecular dynamics simulations to predict binding pockets
Protein interaction studies:
Conduct pulldown assays with cell lysates to identify binding partners
Perform yeast two-hybrid screening
Use protein arrays to identify possible interactions
Validate interactions with co-immunoprecipitation
Activity screening:
Test for enzymatic activities common to bacterial proteins
Assess effects on cell signaling pathways
Examine impacts on bacterial physiology when overexpressed
Loss-of-function studies:
Create knockout strains using CRISPR-Cas systems
Perform phenotypic characterization under various growth conditions
Conduct transcriptome analysis to identify affected pathways
Localization studies:
Use fluorescence microscopy with GFP-lp_2507 fusions
Perform subcellular fractionation to determine natural localization
Assess temporal expression patterns during bacterial growth phases
The combination of these approaches can provide converging evidence about the function of this previously uncharacterized protein.
When encountering low expression yields, a systematic approach is necessary:
Specific recommendations for lp_2507:
Consider using a truncated anchor system (pgsA') which has shown improved expression compared to full-length systems
Empirically determine optimal induction parameters (time, temperature, inducer concentration)
Monitor protein expression at different growth phases to identify ideal harvest points
This distinction is critical for accurate characterization:
Tag-based differentiation:
Express recombinant lp_2507 with affinity tags (His, FLAG, HA)
Use tag-specific antibodies for selective detection
Employ tag-based purification to isolate only recombinant protein
Mass spectrometry approaches:
Compare peptide mass fingerprints between native and recombinant proteins
Identify post-translational modifications that differ
Use stable isotope labeling to differentiate recombinant protein
Specific antibody development:
Generate antibodies against unique epitopes present only in recombinant versions
Use epitope mapping to confirm specificity
Develop sandwich ELISA systems with tag-specific capture and protein-specific detection
Expression level analysis:
Compare expression levels in recombinant vs. wild-type strains
Quantitative Western blotting with density analysis
qRT-PCR to distinguish expression levels at mRNA level
These approaches enable researchers to accurately quantify recombinant protein expression and distinguish it from background levels of native protein.
Confirming proper folding and functionality requires multiple complementary approaches:
Structural analysis:
Circular dichroism (CD) spectroscopy to assess secondary structure
Nuclear magnetic resonance (NMR) for structural characterization
X-ray crystallography for high-resolution structure determination
Comparative analysis with computational structure predictions
Functional assays:
Develop activity assays based on predicted function
Compare activity between recombinant and native protein (if available)
Assess dose-response relationships to confirm specific activity
Binding studies:
Surface plasmon resonance (SPR) to measure binding kinetics with potential partners
Isothermal titration calorimetry (ITC) to determine thermodynamic parameters
Microscale thermophoresis for interaction analysis
Stability assessment:
Thermal shift assays to determine melting temperature
Limited proteolysis to assess structural integrity
Size exclusion chromatography to evaluate aggregation state
Cell-based assays:
Compare cellular responses to recombinant vs. native protein
Assess physiological effects in relevant model systems
Analyze signaling pathway activation patterns
The integration of these methods provides a comprehensive assessment of whether recombinant lp_2507 faithfully recapitulates the structure and function of the native protein.
When faced with contradictory results across expression systems:
Systematic comparison framework:
Create a standardized analytical pipeline for all expression systems
Include positive controls that perform consistently across systems
Document all variables between systems (media, growth conditions, extraction methods)
Expression system-specific considerations:
E. coli: Evaluate effects of inclusion body formation and refolding
Yeast/insect cells: Assess impact of different post-translational modifications
Mammalian cells: Consider species-specific chaperone interactions
Analytical approaches to resolve contradictions:
Perform parallel structural analysis (CD spectroscopy, thermal shift) across all versions
Develop multiple independent functional assays to triangulate true activity
Use mass spectrometry to identify system-specific modifications
Biological relevance assessment:
Determine which system best recapitulates the native bacterial environment
Consider the research question when selecting the most appropriate data
Validate key findings in the most relevant model system
Descriptive statistics:
Calculate means, standard deviations, and coefficients of variation
Generate box plots to visualize distribution of expression levels
Identify outliers using established statistical methods (Grubb's test)
Inferential statistics for comparing conditions:
For normally distributed data: t-tests (paired or unpaired) or ANOVA for multiple conditions
For non-normally distributed data: Mann-Whitney U test or Kruskal-Wallis test
Post-hoc tests (Tukey's, Bonferroni) for multiple comparisons
Variance components analysis:
Identify sources of variability (between batches, days, operators)
Perform nested ANOVA to quantify contribution of each factor
Use results to focus optimization efforts on highest-impact variables
Power analysis:
Calculate appropriate sample sizes needed for statistical significance
Determine minimum detectable differences based on observed variability
Adjust experimental design accordingly
Multivariate approaches:
Principal component analysis to identify patterns across multiple parameters
Multiple regression for modeling relationships between expression conditions and yields
Response surface methodology for optimizing multiple parameters simultaneously
These statistical approaches help distinguish meaningful effects from experimental noise and guide efficient optimization of expression conditions.
Rigorous experimental design is crucial for establishing causal relationships:
Controlled comparison framework:
Wild-type L. plantarum vs. lp_2507 knockout strain
Empty vector control vs. lp_2507 overexpression
Dose-response relationships with purified protein
Time-course experiments to distinguish primary from secondary effects
Comprehensive metabolic analysis:
Metabolomics to assess global metabolic changes
Isotope labeling to track specific metabolic pathways
Respirometry to measure oxygen consumption and metabolic activity
Growth curve analysis under various nutrient conditions
Immune response characterization:
Mechanistic validation:
Inhibitor studies to block specific pathways
Genetic knockdowns of putative interaction partners
Rescue experiments to confirm specificity
Structure-function studies with protein variants
Cross-validation in multiple systems:
Different cell types (macrophages, dendritic cells, epithelial cells)
Ex vivo tissue models
In vivo models where appropriate
Comparison with known immunomodulatory proteins as benchmarks