Ribose-phosphate pyrophosphokinase 1 (PRS1), encoded by the PRPS1 gene, catalyzes the synthesis of phosphoribosylpyrophosphate (PRPP), a critical precursor for nucleotide biosynthesis. In humans, PRS1 is essential for purine, pyrimidine, and tryptophan metabolism . Recombinant expression in L. plantarum would leverage its capacity to produce PRPP for industrial or therapeutic applications, such as nucleoside analog production or metabolic engineering.
L. plantarum is widely used for heterologous protein production due to its food-grade status, robustness, and genetic tractability. Key features include:
Secretion Systems: Utilizes Sec-dependent pathways for extracellular protein release, with signal peptides enabling efficient translocation .
Expression Vectors: pSIP systems (e.g., pSIP401) allow inducible expression via nisin-controlled promoters .
Protein Yield: Achieves high secretion efficiency (e.g., 91% for α-amylase) , though yields vary by target protein and signal peptide design.
While no direct studies on L. plantarum PRS1 exist, lessons from analogous systems highlight potential hurdles:
Cytosolic Localization: PRS1 functions intracellularly, requiring cytoplasmic expression strategies.
Metabolic Burden: Overexpression of PRPP-synthetic enzymes may disrupt cellular nucleotide pools, necessitating strain optimization .
Plasmid Stability: Replication origin compatibility and antibiotic resistance markers (e.g., ermB) influence long-term expression stability .
Recent advancements in L. plantarum genetic engineering include:
Phage-Derived Promoters: Novel inducible systems with up to 9-fold higher expression than native promoters .
CRISPR-Cas9 Systems: Enables precise gene knockouts or insertions for strain optimization .
Quorum Sensing Regulators: pln operons modulate bacteriocin production and stress responses, potentially applicable to PRS1 regulation .
Recombinant PRS1 in L. plantarum could address:
Nucleotide Biosynthesis: Enhanced PRPP production for biotechnological nucleoside synthesis.
Probiotic Metabolic Engineering: Tailoring metabolic pathways for gut microbiota modulation, as seen with butyrate-producing strains .
Therapeutic Delivery: Leveraging L. plantarum’s mucosal adjuvant properties to deliver PRS1-derived antigens or enzymes .
PRS1 Ortholog Identification: No L. plantarum PRS1 homolog is annotated in public databases; ortholog screening (e.g., E. faecium RseP in ) may identify functional candidates.
Metabolic Flux Analysis: Quantifying PRPP pools and nucleotide precursor flux post-PRS1 overexpression.
In Vivo Stability: Assessing recombinant strain persistence in gastrointestinal environments.
This protein is involved in the biosynthesis of the central metabolite phospho-α-D-ribosyl-1-pyrophosphate (PRPP). This occurs through the transfer of a pyrophosphoryl group from ATP to the 1-hydroxyl group of ribose-5-phosphate (Rib-5-P).
KEGG: lpl:lp_0471
STRING: 220668.lp_0471
Ribose-phosphate pyrophosphokinase 1 (PRPS1/prs1) is an essential enzyme that catalyzes the phosphoribosylation of ribose 5-phosphate to 5-phosphoribosyl-1-pyrophosphate (PRPP). This reaction is a critical step in the de novo pathway for purine, pyrimidine, histidine, and tryptophan biosynthesis . The enzyme functions at a key metabolic intersection, connecting the pentose phosphate pathway with nucleotide synthesis pathways. Research has demonstrated that PRPS1 is expressed across various developmental stages in organisms and plays fundamental roles in cellular metabolism by providing the PRPP substrate necessary for nucleic acid synthesis .
The catalytic activity of PRPS1 makes it particularly important in rapidly dividing cells where nucleotide demand is high. In eukaryotic systems, PRPS1 has been associated with various physiological processes beyond basic metabolism, including potential roles in resistance mechanisms to environmental stressors, as evidenced by studies in insect models .
Lactobacillus plantarum has emerged as a valuable expression system for recombinant proteins due to several advantageous characteristics:
Mucosal delivery capability: L. plantarum can survive passage through the gastrointestinal tract, making it an excellent vector for delivering proteins to mucosal surfaces where they can stimulate local immune responses .
Safety profile: L. plantarum has Generally Recognized As Safe (GRAS) status, minimizing regulatory concerns for research applications.
Immunomodulatory properties: Beyond serving as a protein delivery vehicle, L. plantarum itself possesses immunomodulatory properties that can enhance immune responses to recombinant antigens .
Versatile expression systems: Multiple genetic tools and vector systems have been developed specifically for L. plantarum, facilitating the expression of heterologous proteins.
Studies have demonstrated successful application of recombinant L. plantarum for expressing various antigens, including viral components like influenza virus antigen HA1, making it particularly valuable for vaccine research and immunological studies .
Researchers working with L. plantarum typically utilize several established expression vector systems, each with distinct advantages depending on research objectives:
pSIP vectors: These inducible expression vectors contain signal peptides for protein secretion and are regulated by a quorum-sensing mechanism, allowing controlled expression.
pWCF vectors: As demonstrated in research with influenza virus antigens, pWCF vectors can be effectively used for surface display or secretion of heterologous proteins in L. plantarum .
pIB/V5-His expression vectors: While primarily designed for insect cell expression, modified versions have been adapted for lactic acid bacteria including L. plantarum, providing options for His-tagged protein expression and simplified purification .
When selecting an appropriate vector, researchers should consider:
Required expression levels (constitutive vs. inducible)
Protein localization needs (intracellular, secreted, or surface-anchored)
The presence of affinity tags for downstream purification
Antibiotic resistance markers compatible with L. plantarum
The choice of vector significantly impacts expression efficiency, protein localization, and ultimately the success of the recombinant protein production strategy.
Optimizing prs1 expression in L. plantarum requires a multifaceted approach addressing several key factors:
Codon Optimization Strategies:
Adapting the prs1 sequence to L. plantarum codon usage bias significantly improves translation efficiency. Studies with other recombinant proteins in lactic acid bacteria have shown 2-10 fold expression increases following codon optimization. Key considerations include:
Avoiding rare codons in L. plantarum
Optimizing the 5' region of the coding sequence
Eliminating internal Shine-Dalgarno-like sequences that may interfere with translation
Promoter Selection:
The choice of promoter dramatically affects expression levels. Options include:
Constitutive promoters (P23, P59) - providing continuous expression
Inducible systems (nisin-controlled, sakacin-regulated) - allowing controlled expression timing
Stress-responsive promoters - enabling environment-dependent expression
Signal Peptide Optimization:
For secreted prs1 constructs, signal peptide selection affects secretion efficiency. Common signal peptides used with L. plantarum include:
Usp45 (from L. lactis)
SlpA (from L. brevis)
Lp_0373 (native to L. plantarum)
Experimental data from expression studies with similar-sized proteins suggest that Usp45 typically yields 1.5-3 times higher secretion efficiency compared to other signal peptides in L. plantarum systems .
Assessment of immune responses to recombinant L. plantarum requires comprehensive evaluation across multiple immune parameters:
Humoral Immune Response Assessment:
Quantification of serum antibodies (IgG, IgG1, IgG2a) through ELISA
Functional antibody assays such as hemagglutination inhibition (HI) for influenza antigens
B-cell population analysis in lymphoid tissues via flow cytometry (B220+IgA+ cells)
Cellular Immune Response Evaluation:
Flow cytometric analysis of T-cell subsets (CD4+IFN-γ+ and CD8+IFN-γ+ cells)
Lymphocyte proliferation assays upon antigen stimulation
Cytokine profiling using ELISA or intracellular cytokine staining
Mucosal Immunity Assessment:
Secretory IgA measurement in mucosal secretions (intestinal, respiratory)
Analysis of immune cell populations in mucosal-associated lymphoid tissues
Research with recombinant L. plantarum expressing influenza antigens demonstrated significant increases in:
CD4+IFN-γ+ and CD8+IFN-γ+ cells in spleen and mesenteric lymph nodes
B220+IgA+ cells in Peyer's patches
IgA levels in lung and intestinal tissues
These comprehensive assessment approaches provide insight into the multifaceted immune responses generated by recombinant L. plantarum constructs.
PRPS1/prs1 functions at a critical metabolic junction, and its expression level can significantly impact nucleotide metabolism in bacterial systems:
Metabolic Impact of prs1 Expression:
PRPS1 catalyzes the synthesis of phosphoribosylpyrophosphate (PRPP), which serves as an essential precursor for:
De novo purine synthesis
De novo pyrimidine synthesis
Histidine biosynthesis
When prs1 is overexpressed in bacterial systems, several effects may occur:
The ramifications of altered PRPP synthesis extend beyond simple nucleotide availability, potentially affecting:
Cell wall synthesis through pentose phosphate pathway intermediates
Energy metabolism through ATP consumption in PRPP synthesis
Amino acid metabolism through histidine and tryptophan biosynthesis
Research in insect models has suggested that increased PRPS1 expression can support enhanced transcriptional responses, which may similarly apply to bacterial systems with elevated prs1 expression .
Successful cloning of prs1 into L. plantarum expression vectors requires careful attention to several technical aspects:
PCR Amplification Strategy:
For efficient amplification of prs1, consider the following approach:
Design primers with appropriate restriction sites compatible with the target vector
Include a Kozak-like sequence (GAGATGG) before the start codon to enhance translation efficiency
Consider adding two additional nucleotides after the Kozak sequence to maintain the reading frame
Remove the stop codon if C-terminal fusion is planned
A successful primer design strategy based on similar gene cloning would include:
Forward primer: 5′-GGACTAGTGAGATGGAAATG[prs1-specific sequence]-3′ (with SpeI site)
Reverse primer: 5′-CCCTCGAG[prs1-specific sequence without stop codon]-3′ (with XhoI site)
Cloning Procedure:
Digest both PCR product and vector with appropriate restriction enzymes (e.g., SpeI and XhoI)
Purify digested products to remove enzymes and buffer components
Ligate using T4 DNA ligase with an optimal insert:vector ratio (typically 3:1 to 5:1)
Transform into an appropriate E. coli strain (e.g., TOP10) for plasmid propagation
Screen transformants by colony PCR using vector-specific and gene-specific primers
Sequence verify positive clones before transformation into L. plantarum
For vector selection, consider plasmids with strong constitutive promoters for metabolic enzymes like prs1, unless regulated expression is specifically required.
Accurate quantification of prs1 expression levels is crucial for experimental consistency and data interpretation:
Transcript Level Quantification:
Real-time quantitative PCR (qPCR) provides precise measurement of prs1 mRNA levels:
Extract total RNA using specialized kits for Gram-positive bacteria
Synthesize cDNA using reverse transcriptase
Perform qPCR with prs1-specific primers (product size ~150-200 bp is optimal)
Normalize against a reference gene (β-actin or 16S rRNA) using the 2^(-ΔΔCT) method
Example primer design for prs1 qPCR:
Protein Level Quantification:
Western blotting provides direct measurement of PRPS1 protein:
Extract total protein from L. plantarum cultures
Perform SDS-PAGE separation
Transfer to membrane and probe with:
Anti-PRPS1 antibodies (if available)
Anti-tag antibodies (if fusion protein)
Quantify band intensity relative to a loading control
Enzyme Activity Assay:
Functional quantification through PRPS1 enzyme activity:
Prepare cell-free extracts from recombinant L. plantarum
Measure PRPP formation through coupled enzymatic assays
Calculate specific activity (μmol PRPP formed/min/mg protein)
This multi-level quantification approach (transcript, protein, activity) provides comprehensive assessment of prs1 expression and functionality in the recombinant system.
Expressing metabolic enzymes like prs1 in bacterial systems can create significant metabolic burdens that must be carefully managed:
Expression System Optimization:
Inducible promoters: Utilize inducible systems (nisin, sakacin) rather than constitutive promoters to control expression timing and level
Balanced plasmid copy number: Choose medium to low copy number vectors to prevent excessive gene dosage
Codon harmonization: Adjust codon usage to match L. plantarum's natural translation rhythm rather than simply using the most frequent codons
Culture Condition Strategies:
Two-phase cultivation:
Initial growth phase: Focus on biomass accumulation (no induction)
Production phase: Induce expression under optimized conditions
Nutrient supplementation: Provide additional nucleotide precursors to support increased PRPP demand:
Ribose or ribose-5-phosphate
Adenine and guanine bases
Amino acid supplements (particularly histidine and tryptophan)
Environmental parameters:
Temperature reduction during expression phase (25-30°C)
pH control within optimal range for L. plantarum (5.5-6.5)
Controlled oxygen levels based on expression requirements
Genetic Background Considerations:
Consider using L. plantarum strains with enhanced metabolic capacity or deleted competing pathways that might divert metabolic flux away from desired processes.
These strategies can be implemented individually or in combination to minimize metabolic burden while maintaining adequate prs1 expression levels.
Recombinant L. plantarum expressing prs1 presents several valuable applications in metabolic engineering:
Nucleotide Production Enhancement:
By overexpressing prs1, researchers can create strains with enhanced nucleotide biosynthetic capacity, useful for:
Improved DNA/RNA yield in molecular biology applications
Enhanced nucleotide-derived metabolite production
Increased resistance to nucleotide synthesis inhibitors
Metabolic Flux Redirection:
Modulating PRPP availability through prs1 expression can redirect carbon flux through different metabolic pathways:
Increased flux toward nucleotide biosynthesis
Enhanced production of histidine and tryptophan
Altered pentose phosphate pathway utilization
Growth Optimization in Nutrient-Limited Conditions:
Enhanced prs1 expression can potentially improve growth under:
Purine or pyrimidine limitation
Amino acid-restricted conditions
Environments requiring rapid adaptation to metabolic stress
Potential Applications in Resistance Studies:
Based on findings in insect models, where increased PRPS1 expression was associated with deltamethrin resistance, engineered L. plantarum could serve as a model system for studying metabolic aspects of resistance mechanisms . The enhanced nucleotide synthesis capacity may support upregulation of detoxification enzymes and stress response proteins.
Maintaining stable expression of heterologous genes like prs1 in L. plantarum presents several challenges that researchers must address:
Genetic Stability Challenges:
Plasmid Loss Without Selection Pressure:
Plasmids carrying prs1 may be lost over multiple generations without continuous selective pressure
Strategies to address: Implement post-segregational killing systems or balanced-lethal systems for plasmid maintenance without antibiotics
Recombination and Genetic Rearrangements:
Homologous regions may lead to recombination events
Direct repeats in expression cassettes can result in deletion of the inserted gene
Mitigation: Minimize sequence repeats and monitor genetic stability through regular sequencing
Metabolic Burden Leading to Suppressor Mutations:
High prs1 expression may select for mutations reducing expression
Consequences: Gradual decline in expression levels over generations
Solution: Moderate expression levels and regular assessment of expression stability
Monitoring Strategies:
| Generation | Recommended Stability Monitoring Approaches |
|---|---|
| Early (1-5) | - Restriction analysis of extracted plasmids - Colony PCR screening - Expression analysis by Western blot |
| Middle (5-15) | - Sequencing of the expression cassette - qPCR for copy number determination - Enzyme activity assays |
| Late (>15) | - Whole plasmid sequencing - Comparative expression analysis - Growth rate and metabolic profiling |
Stabilization Approaches:
Chromosomal Integration:
Consider integrating prs1 into the L. plantarum chromosome for enhanced stability, though typically at lower expression levels than plasmid-based systems
Selective Medium Development:
Design cultivation media that provide selective advantage to prs1-expressing cells, potentially leveraging the metabolic effects of enhanced PRPP production
Strain Engineering:
Develop host strains with reduced homologous recombination capacity or engineered dependency on the prs1 expression system
These approaches can be combined to develop stable expression systems suitable for long-term studies or applications of recombinant L. plantarum expressing prs1.
When encountering inconsistent results in prs1 expression studies, a systematic troubleshooting approach is essential:
Experimental Variability Assessment:
Expression Level Verification:
Quantify prs1 expression across experimental replicates using qPCR and Western blotting
Determine whether inconsistencies correlate with expression level variations
Standardize induction parameters and harvest timing to minimize variability
Growth Condition Documentation:
Maintain detailed records of media composition, including lot numbers
Monitor and record growth parameters (OD, pH, temperature) throughout experiments
Standardize inoculum preparation and growth phase at harvest
Strain Stability Verification:
Sequence verify plasmids from inconsistent experimental samples
Screen for spontaneous mutations affecting expression or function
Assess plasmid copy number variation across replicates
Data Analysis Approaches for Conflicting Results:
Multivariate Analysis:
Apply principal component analysis (PCA) or partial least squares discriminant analysis (PLS-DA) to identify variables contributing to experimental inconsistency
Meta-Analysis Techniques:
Forest plots to visualize effect sizes across experiments
Random-effects models to account for between-study heterogeneity
Subgroup analysis to identify conditions under which effects are consistent
Bayesian Analysis Framework:
Implement Bayesian approaches to incorporate prior knowledge and uncertainty when interpreting contradictory results
Resolution Strategies:
Independent Validation:
Develop alternative assays measuring the same parameters
Use complementary techniques to verify observations
Consider collaborations for external validation
Hypothesis Refinement:
Revisit initial assumptions about prs1 function
Consider context-dependent effects based on metabolic state
Develop testable models explaining apparently contradictory results
This structured approach enables researchers to distinguish between technical variability and genuine biological phenomena when studying complex metabolic effects of prs1 expression.
Statistical analysis of prs1 expression data requires consideration of the biological variability inherent in bacterial cultures:
Recommended Statistical Frameworks:
Data Normalization Approaches:
Statistical Tests for Comparative Analysis:
Student's t-test for simple two-group comparisons with normally distributed data
ANOVA with appropriate post-hoc tests for multi-group comparisons
Non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) for non-normal distributions
Advanced Modeling Approaches:
Mixed-effects models to account for batch variation and repeated measures
Regression analysis to identify factors influencing expression levels
Time-series analysis for expression dynamics over growth periods
Sample Size and Power Considerations:
| Statistical Power | Minimum Biological Replicates | Technical Replicates per Sample | Detectable Fold Change |
|---|---|---|---|
| 80% | 3 | 3 | ≥2.0 |
| 90% | 4 | 3 | ≥1.8 |
| 95% | 5 | 3 | ≥1.5 |
Practical Implementation:
Software Recommendations:
R with specialized packages (limma, DESeq2, edgeR) for expression data
GraphPad Prism for accessible statistical analysis and visualization
SPSS or SAS for complex experimental designs
Reporting Standards:
Always include measures of central tendency AND dispersion (mean ± SD or median with IQR)
Report exact p-values rather than significance thresholds
Provide transparency about outlier handling and data exclusions
Visualization Approaches:
Box plots showing distribution characteristics
Scatter plots with individual data points for small sample sizes
Heat maps for visualizing patterns across multiple experimental conditions
The investigation of recombinant L. plantarum expressing prs1 opens several promising research avenues:
Metabolic Engineering Applications:
Development of L. plantarum strains with enhanced nucleotide production capacity for biotechnological applications
Creation of probiotic strains with improved stress resistance through modulated nucleotide metabolism
Engineering strains with enhanced capacity for synthesis of secondary metabolites dependent on PRPP availability
Immunological Research:
Investigation of potential adjuvant effects associated with altered bacterial metabolism
Studies on how metabolically engineered L. plantarum might influence host-microbe interactions
Development of novel mucosal vaccine delivery systems leveraging metabolically optimized L. plantarum
Fundamental Research:
Elucidation of the complete metabolic impact of prs1 overexpression using systems biology approaches
Comparative studies across different Lactobacillus species to identify species-specific effects
Investigation of potential resistance mechanisms associated with enhanced nucleotide synthesis capacity
Future studies would benefit from integrating multiple omics approaches (transcriptomics, proteomics, metabolomics) to fully characterize the systemic effects of prs1 expression in L. plantarum and its potential applications in biotechnology and health research.
L. plantarum offers distinct advantages and limitations compared to other bacterial expression systems:
Comparative Analysis of Expression Systems:
| Feature | L. plantarum | E. coli | B. subtilis | Other LAB (L. lactis) |
|---|---|---|---|---|
| Safety profile | GRAS status, suitable for in vivo applications | Endotoxin concerns | GRAS status | GRAS status |
| Protein folding | Good for proteins requiring disulfide bonds | Limited disulfide formation | Good folding machinery | Similar to L. plantarum |
| Secretion efficiency | Moderate | Low without engineering | High | Moderate to high |
| Scalability | Moderate | Excellent | Good | Moderate |
| Genetic tools | Developing | Extensive | Well-developed | Intermediate |
| Codon bias | High GC content | Thoroughly characterized | Medium GC content | Similar to L. plantarum |
| Immunomodulatory properties | Significant | Immunogenic | Minimal | Variable |
Unique Advantages of L. plantarum:
Survival through GI tract enabling mucosal delivery applications
Natural immunomodulatory properties that can enhance responses to heterologous antigens
Ability to deliver proteins to mucosal surfaces while minimizing systemic exposure
Potential for use as a live vector in food and feed applications due to GRAS status
Current Limitations:
Lower protein yields compared to industrial E. coli systems
More limited genetic toolkit compared to model organisms
Variable expression levels requiring optimization for each protein
Challenges in large-scale cultivation compared to industrial production strains