Recombinant Lactobacillus plantarum Inosose dehydratase (iolE)

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 fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
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 collect 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%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is crucial for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. To prioritize a specific tag, please inform us during your order placement.
Synonyms
iolE; lp_3607; Inosose dehydratase; EC 4.2.1.44; 2-keto-myo-inositol dehydratase; 2KMI dehydratase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-300
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Lactobacillus plantarum (strain ATCC BAA-793 / NCIMB 8826 / WCFS1)
Target Names
iolE
Target Protein Sequence
MSSKAEKDIK WGIAPIGWRN DDIPSIGKDN NLQQLLSDIV VAGFQGTEVG GFFPGPEKLN YELKLRNLEI AGQWFSSYII RDGIEKASEA FEKHCQYLKA INAPVAVVSE QTYTIQRSDT ANIFKDKPYF TDKEWDEVCK GLNHYGEIAA KYGLKVAYHH HMGTGIQTKE ETDRLMANTD PKLVGLLYDT GHIAVSDGDY MALLNAHIDR VVHVHFKDVR RSKEEECRAK GLTFQGSFLN GMFTVPGDGD LDFKPVYDKL IANNYKGWIV VEAEQDPSKA NPLEMAQIAH RYIKQHLIEN
Uniprot No.

Target Background

Function
Catalyzes the dehydration of inosose (2-keto-myo-inositol, 2KMI, or 2,4,6/3,5-pentahydroxycyclohexanone) to 3D-(3,5/4)-trihydroxycyclohexane-1,2-dione (D-2,3-diketo-4-deoxy-epi-inositol).
Database Links

KEGG: lpl:lp_3607

STRING: 220668.lp_3607

Protein Families
IolE/MocC family

Q&A

What is the role of inosose dehydratase (iolE) in Lactobacillus plantarum metabolism?

Inosose dehydratase (iolE) in L. plantarum plays a crucial role in the inositol metabolism pathway, catalyzing the conversion of inosose to 2-deoxy-5-keto-D-gluconic acid. This enzyme is part of the broader carbohydrate metabolism network that enables L. plantarum to utilize alternative carbon sources in nutrient-limited environments. While the search results don't specifically detail iolE function, related research on L. plantarum's metabolic pathways indicates that these specialized enzymes contribute to the organism's remarkable adaptability across diverse ecological niches .

Similar to the nucleoside hydrolase genes (iunH, yxjA, rihA, and rihC) identified in L. plantarum SQ001, the iolE gene likely participates in specialized metabolic functions that give this bacterium its distinctive physiological capabilities. Research focusing on metabolic enzymes in L. plantarum demonstrates that these bacteria can modify their metabolic activities in response to different substrates, as evidenced by the significant changes observed in intracellular metabolite composition when exposed to different carbon sources .

What are the standard protocols for cloning and expressing the iolE gene in recombinant L. plantarum systems?

The cloning and expression of genes in recombinant L. plantarum systems typically follow established molecular biology workflows with specific adaptations for this gram-positive bacterium. Based on methods used for similar recombinant L. plantarum studies, researchers should consider a multi-step approach. Initial gene isolation involves PCR amplification of the target iolE gene using primers designed based on conserved regions, followed by insertion into appropriate expression vectors designed for L. plantarum .

Expression vectors like pWCF have been successfully employed in L. plantarum studies, as demonstrated in research with the NC8Δ strain expressing viral antigens . For iolE expression, researchers should select vectors containing appropriate promoters that function efficiently in L. plantarum. Transformation protocols typically involve electroporation with specific parameters optimized for L. plantarum cell walls. Expression verification should include both transcript analysis (RT-PCR) and protein detection methods (Western blot or enzyme activity assays) to confirm successful recombinant production .

How can researchers confirm successful functional expression of recombinant iolE in L. plantarum?

Confirming functional expression of recombinant enzymes requires multiple validation approaches. For recombinant iolE in L. plantarum, researchers should implement a tiered verification strategy. First, molecular confirmation of gene insertion and expression using PCR and RT-PCR to verify the presence and transcription of the iolE gene. Second, protein-level confirmation via Western blot analysis using specific antibodies against the recombinant protein or an added epitope tag .

Most critically, functional validation through enzyme activity assays is essential. Similar to approaches used with nucleoside hydrolase genes, researchers can develop assays that measure the conversion of inosose to its metabolic products . Metabolomic approaches comparing wild-type and recombinant strains can provide comprehensive evidence of functional activity, as demonstrated in studies with L. plantarum SQ001 where metabolite profiles clearly showed changes in nucleoside degradation patterns following genetic manipulation . Additionally, heterologous expression systems (such as E. coli) can be used as complementary validation methods, similar to the approach where L. plantarum SQ001 iunH was expressed in E. coli to confirm its nucleoside degradation function .

What are the optimal conditions for maximizing recombinant iolE expression and activity in L. plantarum?

Optimizing recombinant iolE expression and activity requires systematic evaluation of multiple parameters. Temperature, pH, media composition, and induction timing significantly impact recombinant protein production in L. plantarum. Research with similar recombinant L. plantarum strains suggests cultivation at temperatures between 30-37°C, with slight variations depending on the specific protein's stability characteristics .

Media optimization should focus on balancing growth requirements with expression efficiency. MRS (de Man, Rogosa and Sharpe) medium, commonly used for lactobacilli cultivation, can be modified with carbon source variations to enhance expression. Induction timing is critical - studies with recombinant L. plantarum strains indicate that induction during early-to-mid logarithmic phase often yields optimal results . For iolE specifically, supplementation with inositol or related pathway precursors may enhance expression through metabolic priming.

A systematic factorial design approach is recommended to identify optimal conditions, testing combinations of:

  • Temperature (28°C, 30°C, 37°C)

  • pH (5.5, 6.0, 6.5, 7.0)

  • Carbon source variations (glucose, fructose, inositol supplementation)

  • Induction timing (early, mid, late log phase)

Activity assays should be conducted under various conditions (pH 4.0-8.0, temperatures 25-45°C) to determine the recombinant enzyme's optimal functional parameters, which may differ from expression conditions.

How can researchers design gene knockout experiments to validate the specific metabolic role of iolE in L. plantarum?

Gene knockout experiments provide critical insights into enzyme function within metabolic networks. For validating iolE's role in L. plantarum, researchers should implement both knockout and complementation approaches to establish causality. Based on successful approaches for other genes in L. plantarum, the CRISPR-Cas9 system offers an efficient method for generating precise knockouts .

The experimental design should include:

  • Design of guide RNAs targeting the iolE gene with minimal off-target effects

  • Construction of a CRISPR-Cas9 vector system adapted for L. plantarum

  • Transformation and selection of knockout candidates

  • Molecular verification using PCR and sequencing

  • Phenotypic characterization comparing wild-type and ΔiolE strains

The methodology demonstrated with iunH gene knockout in L. plantarum SQ001, where the strain exhibited approximately 50% reduction in nucleoside degradation capacity following targeted gene deletion, serves as an excellent model . Following knockout verification, comprehensive metabolomic analysis should be performed to identify metabolic bottlenecks or accumulating intermediates resulting from the enzyme's absence. Growth assays with various carbon sources (particularly inositol) will provide functional evidence of the knockout's impact on metabolic capabilities.

Complementation studies, reintroducing the iolE gene on an expression vector, should restore the lost function, thus confirming the direct relationship between the gene and the observed phenotype. This complete knockout-complementation cycle provides the strongest evidence for gene function.

How does recombinant iolE expression affect other metabolic pathways in L. plantarum?

Recombinant enzyme expression can create ripple effects throughout bacterial metabolism due to altered flux distributions, competition for cellular resources, and regulatory responses. For iolE expression in L. plantarum, researchers should anticipate several potential cross-pathway interactions. Overexpression of iolE may increase flux through the inositol degradation pathway, potentially redirecting carbon flow from central metabolism and affecting growth characteristics under different nutrient conditions .

Metabolomic studies similar to those conducted with L. plantarum SQ001 would likely reveal changes extending beyond the immediate inositol pathway. In that research, metabolomic heatmaps showed significant alterations in multiple metabolites following genetic manipulation, including changes in nucleoside levels, nucleobases, and even seemingly unrelated compounds like alpha-linolenic acid and proline . This suggests that iolE overexpression might similarly affect apparently unrelated metabolic pathways through complex regulatory networks.

The carbon flux redistribution resulting from enhanced iolE activity could affect the NAD+/NADH ratio, potentially impacting other redox-dependent pathways. Additionally, competition for transcriptional and translational resources may downregulate other metabolic functions. Comprehensive transcriptomic analysis paired with metabolomics would provide the most complete picture of these complex interactions.

What are the methodological challenges in measuring iolE enzyme kinetics in recombinant L. plantarum systems?

Measuring enzyme kinetics in recombinant bacterial systems presents several methodological challenges specific to the expression system and enzyme characteristics. For recombinant iolE in L. plantarum, researchers must address several key technical considerations.

First, developing a reliable activity assay is essential. Unlike nucleoside hydrolases where substrate degradation can be directly measured as demonstrated in L. plantarum SQ001 research , iolE activity may require more complex detection methods, potentially coupling the reaction to secondary processes that generate detectable products. Spectrophotometric assays monitoring NAD(P)H formation/consumption or chromatographic methods tracking substrate disappearance and product formation are potential approaches.

Second, reliable enzyme extraction and purification protocols must be established. The L. plantarum cell wall presents a barrier to efficient protein extraction, requiring optimization of lysis conditions. Options include enzymatic digestion (lysozyme treatment), mechanical disruption (sonication, bead-beating), or a combination approach. Maintaining enzyme stability during extraction is critical, potentially requiring buffer optimization with stabilizing agents.

Third, distinguishing recombinant iolE activity from native background metabolism requires careful experimental controls. This might include:

  • Using iolE knockout strains as negative controls

  • Employing specific inhibitors if available

  • Creating tagged versions of the enzyme for selective purification

  • Developing differential assay conditions that favor the recombinant enzyme

Finally, enzyme kinetics can be affected by the recombinant context, with potential differences between in vitro measurements and in vivo activity. Researchers should consider both cellular lysate assays and purified enzyme studies to comprehensively characterize the enzyme.

How can researchers integrate computational modeling with experimental data to understand iolE's role in L. plantarum metabolism?

Integrating computational modeling with experimental data creates a powerful approach for understanding enzyme function within complex metabolic networks. For investigating iolE in L. plantarum, researchers should implement a multi-scale modeling approach informed by experimental data at each level.

At the protein level, structural modeling of iolE using homology modeling based on related dehydratases can predict substrate binding sites and catalytic residues. These predictions should guide site-directed mutagenesis experiments to validate computational hypotheses about structure-function relationships. Molecular dynamics simulations can further explore conformational changes during catalysis.

At the pathway level, kinetic modeling of the inositol degradation pathway incorporating experimentally determined parameters (Km, Vmax, substrate/product inhibition) can predict metabolic flux distributions. These models should be validated against experimental metabolomics data, such as the approach used with L. plantarum SQ001 where metabolite levels were quantitatively measured before and after genetic manipulation .

At the genome-scale level, constraint-based metabolic models of L. plantarum can incorporate iolE-specific constraints to predict broader metabolic impacts. These models can generate testable hypotheses about growth phenotypes under various conditions and predict potential metabolic engineering targets to enhance desired outcomes. The metabolomic data collection approach demonstrated with L. plantarum SQ001, using PCA analysis and volcano plots to visualize metabolite changes , provides an excellent framework for generating the experimental data needed to validate such models.

Iterative refinement through cycling between computational predictions and experimental validation will progressively improve model accuracy. This systems biology approach can ultimately reveal how iolE functions within the broader metabolic network of L. plantarum and guide rational engineering of this pathway.

How does the iolE enzyme from L. plantarum compare to homologous enzymes from other bacterial species?

Comparative analysis of iolE across bacterial species provides insights into evolutionary conservation, functional specialization, and potential biotechnological applications. While the search results don't specifically address iolE comparisons, similar approaches to those used in studying other L. plantarum enzymes can be applied.

The inosose dehydratase enzyme belongs to the broader family of sugar dehydratases found across bacterial taxa. Sequence alignment and phylogenetic analysis typically reveal conservation patterns in catalytic domains while showing divergence in regulatory or accessory regions. In Bacillus subtilis, a well-studied model organism for inositol metabolism, the iolE enzyme has been characterized in detail, providing a useful comparison point. L. plantarum's iolE likely shows adaptations reflecting its ecological niche as a lactic acid bacterium found in fermented food environments and the mammalian gut.

Functionally, comparative biochemical characterization between recombinant iolE enzymes from different sources would be expected to reveal variations in:

  • Substrate specificity (strict specificity for inosose vs. activity with related compounds)

  • Kinetic parameters (Km, Vmax, catalytic efficiency)

  • pH and temperature optima

  • Cofactor requirements

  • Allosteric regulation

The methodology for such comparative studies would mirror approaches used in characterizing L. plantarum nucleoside hydrolases, where heterologous expression systems were used to produce and characterize enzymes . This approach allows direct comparison under identical experimental conditions.

What experimental approaches can identify potential regulatory mechanisms controlling iolE expression in L. plantarum?

Understanding the regulatory mechanisms controlling iolE expression requires integrated experimental approaches targeting multiple levels of regulation. Based on studies of other metabolic genes in L. plantarum, several methodological strategies can be applied.

At the transcriptional level, promoter mapping and analysis should be conducted to identify regulatory elements. Reporter gene fusions (using systems like luciferase or fluorescent proteins) can be constructed to monitor promoter activity under different conditions. Growth with various carbon sources (glucose, fructose, inositol) can reveal carbon catabolite repression effects. Similarly, examining expression during different growth phases can identify potential growth-stage dependent regulation .

Transcription factor identification can be approached through:

  • Bioinformatic analysis to identify potential binding sites

  • DNA-protein interaction studies (electrophoretic mobility shift assays)

  • Chromatin immunoprecipitation followed by sequencing (ChIP-seq)

  • Transcription factor knockout studies to identify regulators

Post-transcriptional regulation should also be investigated, including mRNA stability measurements and potential small RNA interactions. At the protein level, post-translational modifications and protein stability under different conditions may provide additional regulatory layers.

The comprehensive approach used in L. plantarum SQ001 metabolic studies, combining genetic manipulation with metabolomic analysis , provides a framework for connecting regulatory mechanisms to functional outcomes. By applying these techniques specifically to iolE, researchers can construct a complete regulatory model of how L. plantarum controls this metabolic function in response to environmental conditions.

How can researchers design experiments to investigate potential horizontal gene transfer of iolE among Lactobacillus species?

Investigating horizontal gene transfer (HGT) of metabolic genes like iolE requires a combination of computational and experimental approaches. While the search results don't directly address HGT studies for iolE, similar methodologies can be applied based on microbial genetics principles.

Computational detection of potential HGT events would begin with comprehensive phylogenetic analysis of iolE sequences across Lactobacillus species and related genera. Incongruencies between gene trees and species trees often indicate HGT events. Additional signatures include:

  • Unusual GC content or codon usage patterns in the iolE gene compared to the genome average

  • Presence of mobile genetic elements or integration sites near the iolE gene

  • Sporadic distribution of iolE among closely related species

Experimental approaches to investigate HGT potential include:

  • Laboratory evolution experiments exposing Lactobacillus species lacking iolE to environments where the gene would confer adaptive advantage, followed by genomic analysis to detect acquisition events

  • Conjugation experiments using marked iolE genes to track transfer between Lactobacillus strains under various conditions, similar to methodologies used in tracking plasmid transfer

  • Transformation studies assessing the natural competence of Lactobacillus species for uptake of iolE-containing DNA fragments

  • Molecular epidemiology surveys examining iolE sequence conservation across Lactobacillus isolates from diverse environmental sources to identify potential transfer patterns

These approaches would provide insights into not only the evolutionary history of iolE in Lactobacillus species but also the potential for future spread of metabolic capabilities through microbial communities.

What methodological approaches can optimize recombinant L. plantarum-iolE for potential biotechnological applications?

Optimizing recombinant L. plantarum expressing iolE for biotechnological applications requires systematic engineering at multiple levels. Drawing from successful approaches with other recombinant L. plantarum systems, researchers should consider several key optimization strategies.

Genetic optimization should focus on expression system refinement. Promoter engineering, as demonstrated in studies with recombinant L. plantarum expressing viral antigens , can significantly impact protein production levels. Codon optimization of the iolE gene based on L. plantarum's preferred codon usage can enhance translation efficiency. Additionally, signal peptide optimization for proteins requiring secretion or particular subcellular localization may be necessary depending on the intended application.

Process optimization should address cultivation conditions. A design-of-experiments approach testing various media formulations, feeding strategies, and physical parameters (pH, temperature, oxygen levels) can identify optimal production conditions. Scale-up considerations should be addressed early, moving from shake flask to bioreactor studies with appropriate control parameters.

The functionality of the recombinant system requires validation under application-specific conditions. For iolE-based applications, this might include stability studies under process-relevant conditions, activity assays with actual industrial substrates rather than model compounds, and long-term performance evaluation. The approach used to validate nucleoside degradation in L. plantarum SQ001 through heterologous expression and gene knockout studies provides a model for functional validation .

How can metabolomic approaches be used to troubleshoot unexpected outcomes in recombinant L. plantarum-iolE research?

Metabolomics offers powerful tools for diagnosing and resolving unexpected outcomes in recombinant enzyme research. When recombinant L. plantarum-iolE systems exhibit unpredicted behaviors, a systematic metabolomic investigation can identify underlying causes and guide corrective strategies.

Common issues revealed through metabolomics include:

  • Precursor limitations: Identified by depletion of pathway intermediates upstream of iolE

  • Product inhibition: Revealed by accumulation of pathway products with corresponding decrease in enzyme activity

  • Cofactor imbalances: Detected through altered ratios of redox cofactors or energy carriers

  • Unexpected pathway crosstalk: Shown by changes in seemingly unrelated metabolic pathways

The visualization approaches used in L. plantarum SQ001 research, including PCA, volcano plots, and metabolite heatmaps , are particularly effective for identifying patterns in complex metabolomic data. Once specific metabolic issues are identified, targeted interventions can be designed, such as supplementing limiting precursors, implementing product removal strategies, or engineering additional pathways to resolve bottlenecks.

Time-course metabolomics is especially valuable for troubleshooting, as it can reveal when and how metabolic imbalances develop during cultivation. This temporal information often provides critical clues about the underlying mechanisms of observed issues.

What experimental designs can evaluate the potential of recombinant L. plantarum-iolE for metabolic engineering of inositol utilization pathways?

Evaluating recombinant L. plantarum-iolE for metabolic engineering applications requires systematic experimental approaches that assess both the immediate enzymatic function and broader metabolic impacts. Based on successful metabolic engineering strategies applied to other pathways, researchers should implement a multi-phase experimental design.

Initial strain characterization should establish baseline performance metrics:

  • Growth profiling using inositol as sole carbon source, measuring growth rates, yields, and inositol consumption rates

  • Enzymatic activity measurements comparing wild-type and recombinant strains

  • Metabolomic analysis of pathway intermediates during inositol utilization

Following baseline establishment, pathway optimization experiments should focus on:

  • Overexpression of iolE combined with other inositol pathway enzymes to identify rate-limiting steps

  • Promoter engineering to achieve coordinated expression of pathway components

  • Knockout or downregulation of competing pathways to channel carbon flux through the target pathway

The experimental approach demonstrated in L. plantarum SQ001 research, where metabolomic analysis revealed significant changes in metabolite composition following genetic manipulation , provides an excellent framework for evaluating pathway engineering outcomes. The use of PCA, volcano plots, and KEGG pathway analysis allowed researchers to visualize complex metabolic shifts resulting from genetic changes .

Advanced experimental designs should include:

  • Feeding studies with labeled inositol (13C) to track carbon flux through engineered pathways

  • Adaptive laboratory evolution to further optimize inositol utilization

  • Integration of heterologous genes to extend the pathway toward valuable products

Performance evaluation should utilize both batch and continuous cultivation modes, with the latter better revealing long-term stability and evolutionary robustness of the engineered system.

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.