Recombinant Lactobacillus plantarum Elongation factor 4 1 (lepA1), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested and agreed upon in advance. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, but this can be adjusted as needed.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
Note: While the tag type is determined during production, please specify your desired tag type for preferential development.
Synonyms
lepA1; lp_2015; Elongation factor 4 1; EF-4 1; EC 3.6.5.n1; Ribosomal back-translocase LepA 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Lactobacillus plantarum (strain ATCC BAA-793 / NCIMB 8826 / WCFS1)
Target Names
lepA1
Uniprot No.

Target Background

Function
This protein is essential for accurate and efficient protein synthesis under specific stress conditions. It functions as a fidelity factor in translation, catalyzing one-codon backward translocation of tRNAs on incorrectly translocated ribosomes. This back-translocation moves from a post-translocation (POST) complex to a pre-translocation (PRE) complex, allowing elongation factor G a second chance for correct tRNA translocation. The protein binds to ribosomes in a GTP-dependent manner.
Database Links

KEGG: lpl:lp_2015

STRING: 220668.lp_2015

Protein Families
TRAFAC class translation factor GTPase superfamily, Classic translation factor GTPase family, LepA subfamily
Subcellular Location
Cell membrane; Peripheral membrane protein; Cytoplasmic side.

Q&A

What is the significance of Elongation factor lepA1 in Lactobacillus plantarum research?

Elongation factor lepA1 functions in protein synthesis in L. plantarum, playing a critical role in translation accuracy and efficiency. Its significance in research stems from both understanding fundamental translation mechanisms and its potential as a target for recombinant expression. When expressed as a recombinant protein, lepA1 can serve as a model for studying protein expression systems in L. plantarum, similar to how other proteins like viral antigens have been successfully expressed in this bacterium. Notably, the expression techniques established with recombinant viral antigens in L. plantarum provide valuable methodological frameworks that can be applied to lepA1 expression .

How does Lactobacillus plantarum compare to other bacterial expression systems for recombinant protein production?

L. plantarum offers several advantages compared to other bacterial expression systems. As a food-grade, generally recognized as safe (GRAS) organism, L. plantarum eliminates concerns about endotoxin production common with E. coli systems. L. plantarum is facultatively anaerobic and mesophilic, allowing for flexible culture conditions . The bacterium demonstrates remarkable stability under various environmental conditions, including high temperature (50°C), low pH (1.5), and presence of bile salts, making it particularly suitable for proteins intended for oral administration or gastrointestinal applications . L. plantarum has been successfully used to express various recombinant proteins including viral antigens such as influenza HA1 and SARS-CoV-2 spike protein with high efficiency and stability .

What are the key considerations for selecting appropriate L. plantarum strains for lepA1 expression?

When selecting L. plantarum strains for lepA1 expression, researchers should consider several factors. First, growth characteristics and genetic tractability are important - strains like NC8Δ have proven amenable to genetic modification and recombinant protein expression . Second, the presence or absence of specific genetic markers affects selection strategies; for example, using alanine racemase (alr) gene deletion strains as hosts provides an antibiotic-free selection system . Third, consider the native protein expression profile to minimize interference with lepA1 expression. Fourth, evaluate strain stability under expected experimental conditions. Finally, assess whether the strain has been previously characterized for recombinant protein expression, as strains with demonstrated success (like L. plantarum Lp18 or NC8Δ) may offer established protocols and predictable results .

What plasmid vectors are most effective for lepA1 expression in L. plantarum?

For effective lepA1 expression in L. plantarum, several plasmid vectors have demonstrated success in similar recombinant protein expressions. The pSIP411 vector has been effectively used for surface expression of proteins in L. plantarum, utilizing inducible promoters controlled by the SppIP peptide inducer . For antibiotic-free selection, E. coli-Lactobacillus shuttle expression vectors employing aspartic acid-β-semialdehyde dehydrogenase (asd) and alanine racemase (alr) genes as screening markers have proven effective, particularly when used with corresponding gene-deficient host strains . The pWCF vector has been successfully employed for surface display using pgsA' as an attachment matrix for target proteins . When selecting a vector for lepA1 expression, consider the promoter strength, induction system, copy number, and whether secretion or surface display is desired.

How should codon optimization be performed for lepA1 expression in L. plantarum?

Codon optimization is crucial for efficient lepA1 expression in L. plantarum as it significantly impacts protein yield. The process should begin with analysis of L. plantarum's codon usage bias, focusing on the specific strain being used (e.g., Lp18 or NC8Δ) . When optimizing the lepA1 gene sequence, replace rare codons with synonymous codons frequently used in highly expressed L. plantarum genes. Additionally, eliminate potential RNA secondary structures, especially in the 5' region, that might impede translation initiation. Avoid internal Shine-Dalgarno-like sequences that could cause ribosomal pausing. Research has demonstrated that codon optimization can substantially enhance expression efficiency in L. plantarum, as shown with viral proteins where optimized codons significantly improved surface expression and antigenicity . Commercial synthesis of the codon-optimized lepA1 gene is recommended for precise implementation of the optimized sequence.

What are the most effective strategies for surface display of lepA1 on L. plantarum?

Surface display of lepA1 on L. plantarum can be achieved through several proven strategies. One effective approach is using polyglutamate synthase A (pgsA) from Bacillus subtilis as a surface display anchor, which has demonstrated success in various recombinant protein expressions . Another strategy involves fusion with endogenous signal peptides, such as signal peptide 1320 (ALX04_001320) from L. plantarum, which effectively targets proteins to the cell surface . For enhanced display efficiency, consider incorporating target peptides like DCpep (FYPSYHSTPQRP) at the C-terminus of lepA1, which has been shown to improve surface targeting and presentation . The expression construct should include appropriate flanking regions for proper membrane insertion and surface anchoring. Verification of surface display should employ multiple techniques, including transmission electron microscopy, indirect immunofluorescence assay, and flow cytometry to quantify display efficiency, which typically ranges from 30-40% of the bacterial population under optimal conditions .

How can protein expression levels of recombinant lepA1 be optimized in L. plantarum?

Optimizing lepA1 expression in L. plantarum requires a multifaceted approach targeting various aspects of the expression system. First, induction conditions significantly impact protein yield; optimal results have been achieved with SppIP inducer at 50 ng/mL concentration, 37°C incubation temperature, and 6-10 hour induction period . Second, growth media composition affects both bacterial growth and protein expression; MRS medium supplemented with specific carbon sources can enhance expression levels. Third, growth phase timing is critical - initiating induction during early to mid-logarithmic phase typically yields better results than stationary phase induction. Fourth, codon optimization as previously discussed is essential for maximizing translation efficiency. Fifth, consider modifying the signal peptide or adding stability-enhancing tags if expression remains suboptimal. Systematic optimization through factorial experimental design is recommended, testing various combinations of these parameters to identify optimal conditions specific to lepA1 expression in your selected L. plantarum strain.

What are the common challenges and solutions in purifying recombinant lepA1 from L. plantarum?

Purification of recombinant lepA1 from L. plantarum presents several challenges. First, cell wall disruption can be difficult due to L. plantarum's robust cell wall; combining enzymatic treatment (lysozyme, mutanolysin) with mechanical disruption (sonication, bead-beating) typically yields better results than either method alone. Second, membrane-associated proteins like surface-displayed lepA1 require effective solubilization; detergents such as Triton X-100 or n-dodecyl β-D-maltoside at 0.5-1% concentration are often effective. Third, lepA1's potential instability during purification can be addressed by including protease inhibitors and maintaining low temperatures throughout the process. Fourth, affinity purification using fusion tags (His-tag, HA-tag) can simplify isolation, though tag placement should be carefully considered to avoid interfering with protein function. Finally, endogenous protein contamination may require additional chromatography steps (ion exchange, size exclusion) for achieving high purity. Western blot analysis using specific antibodies should be employed at each purification stage to track lepA1 recovery and purity.

How can researchers assess the stability and functionality of recombinant lepA1 in L. plantarum?

Assessing stability and functionality of recombinant lepA1 in L. plantarum requires multiple analytical approaches. For thermal stability, expose recombinant bacteria to varying temperatures (37-50°C) for defined periods and quantify remaining lepA1 using Western blot analysis, similar to methods used for other recombinant proteins in L. plantarum that demonstrated stability at 50°C for 20 minutes . pH stability can be evaluated by exposing bacteria to pH ranges from 1.5-7.0, which is particularly relevant for oral administration applications . Functionality of lepA1 should be assessed through GTPase activity assays, as lepA1 is a GTPase involved in translation. Long-term expression stability should be monitored over multiple generations and storage conditions. Additionally, structural integrity can be evaluated using circular dichroism spectroscopy. Resistance to proteolytic degradation in relevant biological environments (e.g., gastrointestinal conditions) provides important information about potential applications. A comprehensive stability profile across these parameters will inform optimal storage, administration, and application conditions for recombinant lepA1-expressing L. plantarum.

How might recombinant L. plantarum expressing lepA1 be used in immunological research?

Recombinant L. plantarum expressing lepA1 offers promising applications in immunological research. As a bacterial elongation factor, lepA1 shares certain conserved domains with pathogenic bacterial species, potentially serving as an immunogenic target for studying cross-protective immune responses. The L. plantarum delivery system can activate dendritic cells in Peyer's patches and increase CD4+IFN-γ+ and CD8+IFN-γ+ cell numbers in the spleen and mesenteric lymph nodes, as demonstrated with other recombinant antigens . This system could be used to study mucosal immunity, as recombinant L. plantarum has shown ability to increase B220+IgA+ cells in Peyer's patches and enhance IgA levels in lungs and intestinal segments . Furthermore, lepA1-expressing L. plantarum could serve as a model for investigating bacterial translational machinery as an immunological target, potentially leading to new vaccine strategies against bacterial pathogens or for understanding autoimmune responses to conserved bacterial proteins.

What experimental design would be optimal for evaluating immune responses to recombinant L. plantarum expressing lepA1?

An optimal experimental design for evaluating immune responses to recombinant L. plantarum expressing lepA1 would include the following components:

Animal model selection:

  • Use 6-8 week old female BALB/c mice (10-12 per group) for consistency with previous studies

  • Include control groups: untreated, empty vector L. plantarum, and positive control (if available)

Immunization protocol:

  • Oral administration of 10^9-10^10 CFU recombinant bacteria per dose

  • Schedule: Days 0, 14, and 28 for primary immunization and boosters

  • Collect samples at days 0, 14, 28, and 42

Immune response evaluation:

  • Humoral immunity: Measure serum IgG, IgG1, and IgG2a antibodies via ELISA

  • Mucosal immunity: Analyze fecal and intestinal IgA levels

  • Cellular immunity: Assess T cell responses through:

    • Flow cytometry analysis of CD4+IFN-γ+ and CD8+IFN-γ+ cells in spleen and MLNs

    • T cell proliferation assays using isolated lymphocytes

  • Dendritic cell activation in Peyer's patches

Functional assays:

  • If applicable, evaluate protective efficacy against bacterial challenge

  • Cytokine profile analysis (IL-4, IL-10, IFN-γ, IL-17) to characterize Th1/Th2/Th17 polarization

This comprehensive design would provide detailed characterization of immune responses similar to those observed with other recombinant antigens expressed in L. plantarum .

What are the technical considerations for designing fusion constructs of lepA1 with other proteins of interest?

Designing fusion constructs of lepA1 with other proteins requires careful technical consideration of multiple factors. First, domain structure analysis is essential - understand the functional domains of lepA1 and ensure fusion sites don't disrupt critical regions. Second, linker selection is crucial; flexible glycine-serine linkers (GGGGS)n provide spatial separation between domains, while rigid helical linkers maintain defined orientation between fusion partners. Third, fusion orientation matters - N-terminal vs. C-terminal fusions of lepA1 will affect both expression and function differently; testing both orientations is advisable. Fourth, potential for protein misfolding increases with fusion complexity; consider incorporating solubility-enhancing tags like thioredoxin if misfolding occurs. Fifth, codon optimization should account for the entire fusion construct, not just individual components. Sixth, incorporate appropriate detection tags (His, HA) positioned to minimize functional interference. Finally, surface display requires careful consideration of transmembrane topology - ensure the fusion construct maintains proper orientation relative to the cell membrane. Experimental validation through Western blotting, activity assays, and structural analysis is essential to confirm proper expression and functionality of the fusion protein .

What is the current methodological standard for analyzing metabolic impacts of lepA1 overexpression in L. plantarum?

The current methodological standard for analyzing metabolic impacts of lepA1 overexpression in L. plantarum involves a multi-omics approach. Transcriptomic analysis using RNA-Seq should compare gene expression profiles between lepA1-overexpressing and control strains, focusing on translation-related genes and stress responses. Proteomic analysis using LC-MS/MS can identify changes in the cellular proteome, particularly in pathways affected by altered translation rates. Metabolomic profiling using GC-MS and LC-MS should measure changes in primary and secondary metabolites, especially those involved in energy metabolism and amino acid utilization. Growth curve analysis under various conditions (different carbon sources, stress conditions) provides functional data on metabolic fitness. Cell morphology and ultrastructure should be examined using microscopy techniques to identify any structural changes resulting from lepA1 overexpression. Additionally, ribosome profiling can directly measure translational impacts, while enzyme activity assays for key metabolic enzymes can detect functional metabolic alterations. This comprehensive approach will provide a detailed understanding of how lepA1 overexpression affects L. plantarum metabolism, similar to methodologies used for other recombinant protein expressions in this organism .

What statistical approaches are most appropriate for analyzing lepA1 expression data across different L. plantarum strains?

For analyzing lepA1 expression data across different L. plantarum strains, several statistical approaches are appropriate depending on the experimental design and data characteristics. For comparing expression levels between multiple strains under single conditions, one-way ANOVA followed by appropriate post-hoc tests (Tukey's HSD for all pairwise comparisons or Dunnett's test when comparing against a control strain) is the standard approach . For experiments with multiple factors (strain type, induction conditions, time points), factorial ANOVA or mixed-effects models should be employed to assess main effects and interactions. When analyzing non-normally distributed data, non-parametric alternatives such as Kruskal-Wallis with Dunn's post-hoc test should be used. For time-course expression data, repeated measures ANOVA or linear mixed models are appropriate. Expression data should be log-transformed when necessary to meet assumptions of normality. Statistical significance should be reported with exact p-values, with thresholds at p < 0.05, p < 0.01, p < 0.001, and p < 0.0001 to indicate different levels of significance . Power analysis should be conducted prior to experimentation to ensure adequate sample sizes for detecting biologically meaningful differences in expression levels.

How should researchers address data inconsistencies when comparing different detection methods for recombinant lepA1?

When addressing data inconsistencies between different detection methods for recombinant lepA1, researchers should implement a systematic approach. First, acknowledge that different detection methods (Western blot, flow cytometry, immunofluorescence, enzymatic assays) measure different aspects of protein expression and may not correlate perfectly. Create a standardization protocol where each method is calibrated using known quantities of purified recombinant lepA1 to generate standard curves. When inconsistencies arise, evaluate method-specific limitations - Western blots provide total protein quantity but may include non-functional protein, while activity assays measure only functional protein. Implement Bland-Altman analysis to quantify the degree of agreement between methods and identify systematic biases. Consider using orthogonal approaches and triangulating results - for example, combining Western blot (protein quantity), GTPase activity assay (functional protein), and structural analysis (correctly folded protein). For surface-displayed lepA1, compare flow cytometry and immunofluorescence microscopy data to resolve discrepancies in detection efficiency. Report all results transparently, including discrepancies, and discuss the biological significance of these differences rather than selectively reporting only consistent data points .

What are the key experimental controls necessary for validating successful recombinant lepA1 expression?

Validating successful recombinant lepA1 expression in L. plantarum requires several critical experimental controls. First, include a negative control consisting of the host L. plantarum strain transformed with an empty vector to establish baseline signals for all detection methods. Second, incorporate a positive control using a well-characterized recombinant protein previously expressed in L. plantarum (such as HA1 or spike protein) to validate the expression system . Third, perform genetic verification through PCR and sequencing to confirm the correct lepA1 sequence in the expression construct. Fourth, include multiple detection methods - Western blot analysis using antibodies against both lepA1 and any fusion tags, complemented by functional assays specific to lepA1's GTPase activity. Fifth, for surface display validation, employ non-permeabilized versus permeabilized immunostaining to differentiate surface from intracellular protein. Sixth, include a dose-response analysis if using an inducible system to demonstrate expression correlation with inducer concentration. Finally, implement temporal controls by measuring expression at multiple time points to capture expression dynamics. These comprehensive controls will provide robust validation of lepA1 expression comparable to standards used in published recombinant protein expressions in L. plantarum .

Control TypePurposeExpected Outcome for Valid Expression
Empty vector L. plantarumEstablish baseline/backgroundMinimal to no lepA1 signal
Known recombinant protein in L. plantarumValidate expression systemPositive signal for control protein
Genetic verification (PCR/sequencing)Confirm construct integrityCorrect lepA1 sequence verified
Uninduced vs. induced culturesVerify induction systemSignificant expression increase after induction
Permeabilized vs. non-permeabilized cellsValidate surface displaySignal in non-permeabilized cells if surface-displayed
Purified recombinant lepA1 standardQuantification referenceConcentration-dependent signal correlation
Host strain stability controlAssess construct stabilityMaintained expression over multiple generations

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.