Note: We will prioritize shipping the format currently in stock. However, if you require a specific format, please specify this in your order notes, and we will fulfill your request to the best of our ability.
Note: All protein shipments include standard blue ice packs. Dry ice shipping is available upon request; please contact us in advance as additional charges will apply.
Tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Recombinant Lactobacillus plantarum Ribonuclease 3 (rnc) is a double-stranded RNA-digesting enzyme involved in processing the primary rRNA transcript to produce the immediate precursors of the large (23S) and small (16S) rRNAs. It also processes certain mRNAs and tRNAs when encoded within the rRNA operon, and processes pre-crRNA and tracrRNA of type II CRISPR loci if present in the organism.
KEGG: lpl:lp_1631
STRING: 220668.lp_1631
RNase III is a global regulator of gene expression that recognizes and cleaves double-stranded RNA (dsRNA) at specific targeted locations. In bacterial systems like L. plantarum, RNase III performs several critical functions:
Processing of precursor ribosomal RNA (pre-rRNA) for maturation
Regulating mRNA stability and degradation
Post-transcriptional regulation of numerous genes, including autoregulation of its own expression
Participation in rRNA quality control and localization to the nucleoid
In L. plantarum specifically, RNase III is likely analogous to those in B. subtilis, working alongside other ribonucleases such as RNase Y and PNPase to coordinate RNA metabolism .
To construct recombinant L. plantarum expressing RNase III, researchers require:
Vector selection: Either high copy number (e.g., pCDLbu-1ΔEc-based plasmids) or low copy number (e.g., p256 origin of replication) plasmid backbones
Promoter selection: Options include:
Ribosomal binding site (RBS) optimization: The distance between the Shine-Dalgarno sequence and start codon significantly impacts expression, with 8 nucleotides often providing optimal levels
Cloning strategy: Typically using restriction enzymes (e.g., EcoRI and EcoRV) to insert the gene into expression vectors
Transformation protocol: Electroporation is commonly used for introducing the construct into L. plantarum
Multiple complementary techniques should be employed:
Western blotting: Using anti-RNase III antibodies to detect the expressed protein from cell lysates. This confirms the protein is being produced at the expected molecular weight (typically 25 kDa for RNase III)
Enzymatic activity assay: Measuring dsRNA cleavage activity using synthetic substrates or known natural substrates like 16S rRNA precursors
Immunofluorescence microscopy: Using antibodies to visualize localization of the expressed protein (particularly useful for surface-displayed constructs)
Flow cytometry: Particularly useful for quantifying expression efficiency when the protein is displayed on the bacterial surface (expecting 35-40% positive cells under optimal conditions)
RNase III complementation test: Introducing the construct into an RNase III-deficient strain (e.g., SK7622) and assessing growth rate recovery (a functional RNase III should restore normal growth)
Optimizing RNase III expression requires fine-tuning at multiple levels:
Transcriptional optimization:
Promoter selection based on desired expression level - P11 provides strong expression while Ptuf promoters offer moderate expression
Using high copy number plasmids (e.g., pCDLbu-1ΔEc) can increase yield up to 2-fold compared to low copy vectors
Translational optimization:
Codon optimization for L. plantarum is crucial for heterologous genes
Optimizing the Shine-Dalgarno sequence to match the consensus sequence (AGGAGG)
Varying the spacing between SDS and start codon, with 8 nucleotides typically yielding maximum expression
For surface display, using appropriate secretion signals and anchoring domains (e.g., LPXTG motif)
Induction conditions:
For inducible systems, optimizing inducer concentration (e.g., 50 ng/mL SppIP)
Determining optimal induction time (typically 6-10 hours post-induction)
Cultivation temperature optimization (generally 37°C for L. plantarum)
A systematic approach combining these strategies while monitoring growth parameters is recommended for optimal expression.
The interplay between gene copy number and promoter strength significantly impacts expression levels:
Gene copy number effects:
High copy number plasmids (e.g., based on the L. buchneri CD034 pCD034-1 origin) can contain >200 copies per chromosome
Low copy number vectors (e.g., p256 origin) maintain fewer plasmid copies
High copy number vectors can increase yield approximately 2-fold when combined with the same promoter
Promoter strength effects:
P11 promoter provides strong constitutive expression
Interaction between variables:
Strong promoters on high copy vectors may cause excessive metabolic burden
For RNase III specifically, overexpression can be detrimental as it may disrupt normal RNA processing
The combination of promoter strength and copy number should be calibrated to the specific application
| Vector Type | Promoter | Relative Expression | Growth Impact |
|---|---|---|---|
| High copy | P11 | Very high | Potential growth retardation |
| High copy | Ptuf | High | Minimal impact |
| Low copy | P11 | Moderate | Minimal impact |
| Low copy | Ptuf | Low | No impact |
For RNase III expression, moderate expression levels are often preferable as they prevent disruption of normal cellular RNA processing while providing sufficient enzyme activity .
Several complementary approaches can be used to measure RNase III activity:
In vitro activity assays:
dsRNA cleavage assay: Using synthetic dsRNA substrates labeled with fluorescent dyes and measuring fluorescence after cleavage
Gel electrophoresis analysis: Incubating purified recombinant RNase III with known substrates and analyzing cleavage patterns by gel electrophoresis
In vivo functional assessment:
Growth rate measurement: RNase III activity correlates with bacterial growth; cells lacking functional RNase III grow approximately 25% slower
rRNA processing analysis: Examining 16S rRNA maturation using Northern blotting or primer extension
Autoregulation analysis: Quantifying the ability of RNase III to regulate its own mRNA through 5' UTR cleavage
Nucleoid localization: Assessing the ability of RNase III to localize pre-16S rRNA to the nucleoid using RNA-FISH (fluorescence in situ hybridization)
Complementation studies:
Transform the recombinant L. plantarum RNase III construct into an RNase III-deficient E. coli strain (e.g., SK7622)
Measure growth rate recovery compared to wild-type strains
Analyze rRNA processing patterns to confirm restoration of RNase III activity
Specific mutations in RNase III can provide valuable experimental tools:
Catalytic domain mutations:
E117K mutation: Creates a nuclease-null variant that maintains RNA binding ability but eliminates catalytic activity. Useful for distinguishing binding from cleavage effects
Mutations in the RNase III signature motif: Residues in this 10-residue motif are critical for enzyme function, with most substitutions causing complete activity loss
RNA binding domain mutations:
Mutations in RBM1 and RBM3: These binding motifs are crucial for dsRNA interaction; mutations here reduce substrate affinity without affecting catalytic capability
RBM4 and RBM2 mutations: These regions tolerate more substitutions, allowing for tunable binding affinity
Experimental applications:
Separating binding from cleavage: Use E117K mutant to study RNA binding effects independent of processing
Conditional activity: Engineer temperature-sensitive RNase III variants for temporal control
Substrate specificity studies: Mutations at G97, G99, and F188 appear to affect substrate selectivity without eliminating general activity
| Mutation Region | Function Impact | Experimental Use |
|---|---|---|
| Active site (E117K) | Eliminates cleavage, preserves binding | Binding studies |
| Signature motif | Complete loss of function | Negative control |
| RBM1/RBM3 | Reduced binding affinity | Tuning substrate preference |
| G97/G99/F188 | Altered substrate specificity | Studying specificity determinants |
These mutations provide powerful tools for dissecting RNase III function in L. plantarum gene regulation networks .
Recombinant L. plantarum expressing RNase III has several potential therapeutic applications:
Autoimmune disease treatment:
In rheumatoid arthritis, regulatory T-cell development and function is regulated by FOXP3, which is underexpressed in patients. L. plantarum could be engineered to express both RNase III and FOXP3 to restore T-cell functionality and decrease inflammation
RNase III could help regulate the processing and stability of the FOXP3 mRNA and protein expression
Vaccine delivery platform:
The RNase III enzyme can be co-expressed with vaccine antigens to modulate immune responses by controlling mRNA stability and antigen expression levels
This approach has shown promise for vaccines against viral pathogens (e.g., influenza) and parasitic diseases (e.g., trichinellosis)
Cancer immunotherapy:
L. plantarum expressing tumor antigens like NY-ESO-1 along with RNase III could enhance antigen presentation and immune response
The regulated expression via RNase III activity enables controlled release of antigens, promoting more effective T-cell responses
For these applications, researchers should consider:
Designing constructs with appropriate secretion signals or surface-display anchors
Optimizing RNase III expression to maintain viable bacteria while achieving therapeutic effect
Validating in appropriate animal models before clinical testing
Recombinant L. plantarum expressing RNase III influences host biology through several mechanisms:
Effects on gut microbial communities:
Recombinant L. plantarum can significantly increase microbial diversity as measured by Shannon-Wiener index
It enhances the species richness (Chao1 index) and alters community structure (beta diversity)
These changes may be partially mediated by RNase III's role in processing bacterial sRNAs that regulate gene expression
Immune response modulation:
Humoral immunity: Increases levels of serum IgG, IgG1, IgG2a, and mucosal secretory IgA (sIgA)
Cellular immunity: Enhances CD4+ T cell and CD8+ T cell responses, particularly IFN-γ producing cells
Dendritic cell activation: Activates dendritic cells in Peyer's patches, crucial for antigen presentation
B cell response: Increases the number of B220+IgA+ cells in Peyer's patches
Experimental data from recombinant L. plantarum studies:
| Immune Parameter | Response to Recombinant L. plantarum | Measurement Method |
|---|---|---|
| Serum IgG | 2-5 fold increase | ELISA |
| Mucosal sIgA | Significant increase in intestine and lungs | ELISA |
| CD4+IFN-γ+ cells | Increased in spleen and mesenteric lymph nodes | Flow cytometry |
| B220+IgA+ cells | Increased in Peyer's patches | Flow cytometry |
The mechanism likely involves RNase III's role in regulating bacterial gene expression and antigen processing, which subsequently affects how the recombinant bacteria interact with host immune cells .
Designing robust in vivo experiments requires careful consideration of multiple factors:
Animal model selection:
Mice: Most commonly used for initial studies; BALB/c mice are frequently employed for immunological studies
Disease-specific models: Consider humanized mice, arthritis models, or other specialized models depending on research focus
Administration protocol:
Route: Oral administration is most common for L. plantarum (gavage or in drinking water)
Dosage: Typically 10^8-10^10 CFU per administration, with optimization needed for each construct
Schedule: Multiple doses are usually required; common schedules include:
Controls and groups:
PBS control (negative)
Empty vector L. plantarum (bacterial backbone control)
Recombinant L. plantarum expressing RNase III
Recombinant L. plantarum expressing target antigen without RNase III
Recombinant L. plantarum expressing both RNase III and target antigen
Commercial vaccine or treatment (positive control, if available)
Assessment timeline and methods:
Sampling timepoints: Pre-immunization, 10 days, 20 days, 30+ days post-immunization
Sample types: Serum, feces, intestinal contents, tissue biopsies
Analyses: Flow cytometry, ELISA, RT-qPCR, histopathology, and challenge studies when applicable
Ethical considerations:
Follow appropriate institutional guidelines for animal research
Use power calculations to determine minimum number of animals needed
To investigate RNase III's regulatory role, a comprehensive experimental approach is required:
Construct design and strains:
Wild-type L. plantarum: Baseline control
RNase III knockout/knockdown: Using CRISPR-Cas9 or antisense RNA approaches
RNase III overexpression strain: Using strong constitutive promoter
Catalytically inactive RNase III (E117K): To distinguish binding from cleavage effects
Experimental approaches:
1. Transcriptome analysis:
RNA-seq comparing wild-type, knockout, and overexpression strains
Focus on changes in mRNA levels and processing patterns
Analyze differential expression under various growth conditions (exponential phase, stationary phase, stress conditions)
2. Direct target identification:
CLIP-seq (Cross-linking immunoprecipitation) to identify RNase III-bound RNAs
RNA-seq with size selection to identify processing intermediates
Northern blotting to confirm specific cleavage patterns for candidate targets
3. Functional validation:
Construct reporter systems (e.g., GFP fusions) with candidate target 5' and 3' UTRs
Site-directed mutagenesis of putative RNase III recognition sites
In vitro cleavage assays with purified RNase III and candidate RNA substrates
4. Physiological impact assessment:
Growth curve analysis under various conditions
Stress response testing (acid, bile, oxidative stress)
Competition assays between wild-type and RNase III variant strains
Data analysis approach:
Identify differentially processed/expressed transcripts
Map RNase III cleavage sites on target RNAs
Correlate RNA structural features with cleavage efficiency
Researchers frequently encounter several challenges when expressing RNase III in L. plantarum:
Cause: Overexpression of RNase III disrupting normal RNA processing
Solutions:
Causes: Poor translation efficiency, protein instability, or toxicity
Solutions:
Causes: Limited substrate access or improper assay conditions
Solutions:
Causes: Metabolic burden, recombination events
Solutions:
Causes: Plasmid loss, variable gene expression
Solutions:
Inconsistent immune responses are a common challenge in animal studies with recombinant L. plantarum. Here's a systematic approach to troubleshooting:
Assessment: Verify viable cell count, expression level, and administration technique
Solutions:
Assessment: Examine variability in animal age, weight, gender, and microbiome
Solutions:
Assessment: Measure fecal shedding of the recombinant bacteria
Solutions:
Assessment: Evaluate timing and methods of immune parameter measurement
Solutions:
Troubleshooting flowchart:
Verify bacterial preparation:
Expression level consistent? → If no: Standardize culture conditions
Viability confirmed? → If no: Adjust preparation protocol
Confirm administration:
Consistent dosing? → If no: Standardize administration
Timing appropriate? → If no: Adjust schedule
Assess colonization:
Bacteria detectable in feces? → If no: Increase dose or frequency
Persistence adequate? → If no: Consider prebiotic supplementation
Evaluate immune measurements:
Multiple parameters assessed? → If no: Expand analysis
Appropriate controls included? → If no: Add necessary controls
Implementing this systematic approach will help identify and address sources of variability in immune responses .
RNase III exhibits both conserved and divergent characteristics across bacterial species, with important implications for recombinant expression:
Structural and functional conservation:
The RNase III signature motif is highly conserved across bacterial species
The enzyme architecture consisting of a catalytic domain and dsRNA binding domain is preserved
Basic catalytic mechanism of dsRNA cleavage is consistent across species
Species-specific differences:
Regulatory differences:
In E. coli, RNase III autoregulates through cleavage of its own 5'-UTR
RNase III activity in E. coli can be modulated by phosphorylation during phage infection
The set of natural targets varies between species, affecting the impact of overexpression
Implications for recombinant expression:
Expression optimization: Species-specific codon usage and regulatory elements must be considered
Activity assessment: Target selection for activity assays should consider species-specific substrates
Functional complementation: E. coli RNase III mutants can be used to test L. plantarum RNase III functionality
Physiological impact: Overexpression effects may differ between species based on the targetome
Heterologous applications: Consider using RNase III from different species if specific substrate preferences are desired
Understanding these species-specific differences enables more effective design of recombinant expression systems and accurate interpretation of experimental results across different bacterial models .
Several innovative applications are emerging for recombinant L. plantarum expressing RNase III:
Precision RNA therapeutics:
Using RNase III expression to selectively degrade specific pathogenic RNAs
Engineering substrate specificity through rational mutation of residues G97, G99, and F188
Developing strain-specific RNA targeting for microbiome modulation
Synthetic biology applications:
Creating tunable gene expression circuits using RNase III processing
Developing post-transcriptional regulatory switches based on conditional RNA cleavage
Designing artificial RNA processing systems for metabolic engineering of L. plantarum
Novel vaccine delivery platforms:
Multi-antigen delivery systems with controlled expression via RNase III processing
SARS-CoV-2 and other respiratory pathogen vaccine candidates utilizing surface-displayed antigens
Cancer vaccine development using tumor-associated antigens with RNase III-regulated expression
Therapeutic microbiota modulation:
Targeted alteration of microbiome community structure for disease treatment
Engineering precise strain-specific RNA interference via RNase III-mediated processing
Developing probiotics with enhanced immune-modulatory properties for inflammatory conditions
Diagnostic applications:
Biosensor development using RNase III-dependent reporter systems
In situ detection of specific bacterial targets in complex microbiomes
Monitoring gut health through engineered sentinel bacteria expressing RNase III-regulated reporters
Challenges and future directions:
Improving strain stability and expression consistency
Developing better methods for in vivo monitoring of RNase III activity
Establishing appropriate regulatory frameworks for clinical applications
Despite significant progress, several important knowledge gaps remain in our understanding of RNase III function in L. plantarum:
Current status: Limited knowledge of natural RNA targets in L. plantarum
Research approaches:
Current status: Poor understanding of how RNase III activity changes under different conditions
Research approaches:
Current status: L. plantarum RNase III structure has not been solved
Research approaches:
Current status: Limited knowledge of subcellular localization and dynamics
Research approaches:
Current status: Unknown impact of L. plantarum RNase III on host interactions
Research approaches:
Proposed integrated research strategy:
Generate comprehensive RNase III variant library in L. plantarum
Perform parallel phenotyping under multiple conditions
Apply multi-omics approaches (transcriptomics, proteomics, structuromics)
Develop computational models to predict RNase III activity
Validate in relevant in vivo models including gut colonization studies