Recombinant Listeria monocytogenes serotype 4b Shikimate dehydrogenase (aroE)

Shipped with Ice Packs
In Stock

Description

Shikimate Dehydrogenase Function and Role

Shikimate dehydrogenase catalyzes the reversible reduction of 3-dehydroshikimate to shikimate, utilizing NADPH as a cofactor . This reaction is part of the shikimate pathway, which is absent in animals, making it a target for developing non-toxic herbicides and antimicrobial agents .

Listeria monocytogenes Serotype 4b

Listeria monocytogenes serotype 4b is a significant human pathogen, often associated with severe foodborne illnesses. It is known for its virulence and ability to cause outbreaks . While Listeria monocytogenes does not naturally utilize the shikimate pathway for aromatic amino acid biosynthesis, genetic engineering could potentially introduce such pathways or enzymes for research or biotechnological purposes.

Potential Applications

  • Biotechnology: Recombinant shikimate dehydrogenase could be used in biotechnological applications, such as producing aromatic compounds or developing novel antimicrobial agents.

  • Vaccine Development: Listeria monocytogenes is being explored as a vaccine vector. Introducing shikimate dehydrogenase could potentially enhance its vaccine properties or serve as a marker for genetic modification.

Challenges

  • Genetic Stability: Ensuring the genetic stability of the recombinant strain is crucial.

  • Regulatory Considerations: Any application involving genetically modified organisms must comply with regulatory frameworks.

Data and Research Findings

Given the lack of specific data on recombinant Listeria monocytogenes serotype 4b shikimate dehydrogenase (aroE), the following table summarizes general information about shikimate dehydrogenase and Listeria monocytogenes:

FeatureShikimate DehydrogenaseListeria monocytogenes Serotype 4b
FunctionCatalyzes reduction of 3-dehydroshikimate to shikimateHuman pathogen causing foodborne illnesses
PathwayPart of the shikimate pathway for aromatic amino acid biosynthesisNot naturally involved in the shikimate pathway
ApplicationsTarget for herbicides and antimicrobialsVaccine vector and biotechnological applications
Genetic ModificationCan be engineered into various hostsPotential for genetic modification for biotechnological purposes

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 consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification 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 pellet the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on several 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 maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is defined during production. If you require a specific tag, please inform us, and we will prioritize its implementation.
Synonyms
aroE; LMOf2365_0520Shikimate dehydrogenase; NADP(+); SDH; EC 1.1.1.25
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-291
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
aroE
Target Protein Sequence
MTNKITERIT GHTELIGLIA TPIRHSLSPT MHNEAFAKLG LDYVYLAFEV GDKELKDVVQ GFRAMNLRGW NVSMPNKTNI HKYLDKLSPA AELVGAVNTV VNDDGVLTGH ITDGTGYMRA LKEAGHDIIG KKMTICGAGG AATAICIQAA LDGVKEISIF NRKDDFYANA EKTVEKINSK TDCKAQLFDI EDHEQLRKEI AESVIFTNAT GVGMKPFEGE TLLPSADMLR PELIVSDVVY KPTKTRLLEI AEEQGCQTLN GLGMMLWQGA KAFEIWTHKE MPVDYIKEIL F
Uniprot No.

Target Background

Function

Shikimate dehydrogenase (AroE) is involved in chorismate biosynthesis, a precursor to aromatic amino acids. It catalyzes the reversible NADPH-dependent reduction of 3-dehydroshikimate (DHSA) to shikimate (SA).

Database Links
Protein Families
Shikimate dehydrogenase family

Q&A

What is Shikimate dehydrogenase (AroE) and what is its role in the metabolism of Listeria monocytogenes serotype 4b?

Shikimate dehydrogenase (AroE) is the fourth enzyme in the seven-enzyme shikimate pathway that catalyzes the sequential conversion of erythrose 4-phosphate and phosphoenolpyruvate to chorismate. Specifically, AroE catalyzes the NADPH-dependent reduction of 3-dehydroshikimate to shikimate, a critical step in this metabolic pathway. The shikimate pathway is essential for the synthesis of aromatic amino acids (phenylalanine, tyrosine, and tryptophan) and other aromatic compounds in bacteria, fungi, and plants, but is notably absent in mammals. In Listeria monocytogenes serotype 4b, AroE exists as a monofunctional enzyme, unlike in fungi where it forms part of a pentafunctional arom enzyme complex or in plants where it exists as a bifunctional enzyme with 3-dehydroquinate dehydratase (AroD) . This pathway is crucial for bacterial survival, particularly for intracellular pathogens like L. monocytogenes that must synthesize essential nutrients during infection.

How does Listeria monocytogenes serotype 4b differ from other serotypes in terms of genetic and virulence characteristics?

Listeria monocytogenes serotype 4b strains predominantly belong to lineage I, which also includes serotypes 1/2b, 3b, 4d, and 4e, and is frequently associated with epidemic human listeriosis. Genetic analysis reveals that serotype 4b lineage I strains react with serotype 4b-, 4d-, and 4e-specific ORF2110 and virulence-specific lmo1134 and lmo2821 primers in PCR assays, whereas serotype 4b lineage III strains consistently test negative for ORF2110 and lmo1134 primers . Southern blot analysis using species-specific lmo0733 and virulence-specific lmo2821 gene probes confirms these distinct genetic profiles among different lineages. Lineage I serotypes, particularly 4b, are significantly overrepresented in epidemic outbreaks of human listeriosis compared to lineage II serotypes (which include 1/2a, 1/2c, 3a, and 3c) and lineage III serotypes (4a and 4c) . These genetic differences likely contribute to enhanced virulence and epidemic potential of serotype 4b strains, making them particularly concerning from a public health perspective.

What is the significance of the shikimate pathway as a target for antimicrobial development against Listeria monocytogenes?

The shikimate pathway represents a particularly promising target for antimicrobial development against Listeria monocytogenes for several key reasons. First, this pathway is completely absent in mammals, providing an excellent opportunity for selective toxicity - inhibitors of the pathway could potentially disrupt bacterial metabolism without affecting host cells. Second, the pathway is essential for bacterial survival, as it produces aromatic amino acids and other critical compounds that bacteria cannot obtain from their environment, especially in the nutrient-limited intracellular niche that L. monocytogenes occupies during infection. The structures of AroE are being used as structural templates for the synthesis of effective inhibitors of the shikimate pathway, demonstrating the practical application of this approach . Additionally, targeting metabolic pathways like the shikimate pathway may be less susceptible to rapid resistance development compared to targeting virulence factors, as mutations that circumvent metabolic inhibition often come with significant fitness costs to the pathogen. This combined with the essential nature of aromatic compounds for bacterial survival makes the shikimate pathway an attractive target for novel therapeutics against L. monocytogenes.

What are the most effective expression systems for producing recombinant Listeria monocytogenes serotype 4b Shikimate dehydrogenase (AroE)?

The most effective expression systems for producing recombinant L. monocytogenes serotype 4b AroE typically utilize Escherichia coli platforms, similar to approaches used for other L. monocytogenes proteins. Based on established protocols for recombinant L. monocytogenes proteins, expression systems should be selected according to the intended research application, as outlined in the following table:

Expression SystemAdvantagesRecommended ApplicationsOptimization Strategies
E. coli BL21(DE3)High yield, economical, simpleBiochemical/structural studiesLower temperature (16-25°C), reduced IPTG (0.1-0.5 mM)
E. coli RosettaAddresses rare codon usageEnhanced expression of proteins with rare codonsCo-expression with chaperones for folding assistance
E. coli Arctic ExpressEnhanced protein folding at low temperaturesProteins prone to inclusion body formationExpression at 10-12°C for 24-48 hours
Cell-free systemsAvoids toxicity issues, rapid productionInitial screening, difficult-to-express proteinsSupplementation with NADPH, molecular chaperones

For optimal expression, the aroE gene should be cloned into a vector with a strong but controllable promoter (such as T7) and include appropriate affinity tags (His-tag or GST-tag) to facilitate purification . When expressing recombinant Listeria proteins, attention to codon optimization may be necessary as Listeria and E. coli have different codon usage patterns. Additionally, incorporating a tobacco etch virus (TEV) protease cleavage site between the tag and protein allows for tag removal after purification, which is crucial for crystallography or other structural studies .

What purification strategies yield the highest purity and activity of recombinant AroE protein?

Purification of recombinant AroE to high purity and activity requires a systematic approach combining multiple chromatographic techniques, buffer optimization, and activity preservation strategies. Based on established purification protocols for similar enzymatic proteins, the following multi-step strategy is recommended:

Purification StepMethodKey ParametersConsiderations
Initial CaptureImmobilized Metal Affinity Chromatography (IMAC)5-20 mM imidazole wash, 250-300 mM imidazole elutionInclude 5-10% glycerol and 1 mM DTT to maintain stability
Intermediate PurificationIon Exchange ChromatographypH buffer 0.5-1 units from AroE pI, gradient elutionSelect cation/anion exchange based on AroE theoretical pI
PolishingSize Exclusion ChromatographyFlow rate 0.5-1 ml/min, buffer with 150 mM NaClRemoves aggregates and oligomers
Optional: Tag RemovalTEV Protease Digestion1:20-1:50 TEV:protein ratio, overnight at 4°CRequired for crystallography studies

Throughout all purification steps, it's crucial to maintain reducing conditions (1-5 mM DTT or β-mercaptoethanol) to protect catalytic cysteine residues and include 50-100 μM NADPH to stabilize the enzyme's active site . Purification should be performed at 4°C with protease inhibitors to prevent degradation. For serotype 4b AroE specifically, buffer screening (varying pH 6.5-8.5 and salt concentration 50-300 mM) should be conducted to identify optimal stability conditions. The final purified protein should achieve >90% purity as assessed by SDS-PAGE and maintain specific activity comparable to that of the native enzyme. Optimal storage conditions typically include flash-freezing in liquid nitrogen with 10-20% glycerol and storage at -80°C to maintain long-term activity.

How can researchers verify the correct folding and activity of purified recombinant AroE?

Verification of correct folding and activity of purified recombinant AroE requires a multi-faceted approach combining biophysical characterization and enzymatic assays. The following comprehensive validation strategy ensures both structural integrity and functional activity:

Validation MethodTechnical ApproachExpected Results for Properly Folded AroETroubleshooting Indicators
Secondary Structure AnalysisCircular Dichroism (CD) SpectroscopyAlpha/beta mixed pattern consistent with AroE family proteinsIrregular spectra suggest misfolding
Thermal StabilityDifferential Scanning Fluorimetry (DSF)Single melting transition, Tm >40°CMultiple transitions suggest heterogeneity or aggregation
Homogeneity AssessmentDynamic Light Scattering (DLS)Monodisperse population, <15% polydispersityHigh polydispersity indicates aggregation
NADPH BindingFluorescence TitrationKd values in μM range, changes in intrinsic fluorescenceWeak or absent binding suggests inactive protein
Enzymatic ActivitySpectrophotometric AssayLinear decrease in A340nm due to NADPH oxidationNon-linear kinetics suggest inhibition or inactivation
Kinetic ParametersMichaelis-Menten AnalysisKm values comparable to literature: 50-200 μM for 3-dehydroshikimate, 10-50 μM for NADPHSignificantly altered Km values indicate structural issues

The enzyme activity assay should be performed at 25°C in 100 mM potassium phosphate buffer (pH 7.0) containing 100 μM NADPH and varying concentrations of 3-dehydroshikimate (10-500 μM). The reaction is monitored by following the decrease in absorbance at 340 nm due to NADPH oxidation . For confirmatory purposes, HPLC-based product formation assays can directly quantify shikimate production. For structural applications, limited proteolysis using trypsin or chymotrypsin can provide additional evidence of proper folding, as well-folded proteins typically show resistance to proteolytic digestion except at exposed flexible regions. These complementary approaches collectively provide robust validation of both the structural integrity and catalytic functionality of the purified recombinant AroE.

What are the key structural features of Shikimate dehydrogenase (AroE) that contribute to its catalytic function?

Shikimate dehydrogenase (AroE) possesses several distinct structural features that are essential for its catalytic function in the NADPH-dependent reduction of 3-dehydroshikimate to shikimate. Based on crystallographic studies of AroE enzymes, the following key structural elements directly contribute to its enzymatic activity:

Structural FeatureDescriptionFunctional Significance
Two-Domain ArchitectureN-terminal nucleotide-binding domain and C-terminal substrate-binding domainCreates interdomain catalytic cleft where reaction occurs
Rossmann FoldCharacteristic βαβαβ motif in N-terminal domainNADPH binding and recognition, determines cofactor specificity
Catalytic Dyad/TriadConserved Lys, Asp/Tyr residuesDirect involvement in proton transfer during catalysis
Substrate-Binding PocketPositively charged pocket with specific hydrogen bonding residuesRecognition and orientation of 3-dehydroshikimate
Interdomain FlexibilityHinge regions allowing domain movementEnables "closed" conformation upon substrate binding
Anion Binding SiteConserved arginine residuesStabilizes carboxylate group of substrate

The AroE enzyme typically undergoes significant conformational changes upon binding of both NADPH and substrate, transitioning from an "open" to a "closed" state that brings catalytic residues into optimal proximity for hydride transfer . The specific binding mode of NADPH positions the pro-4S hydrogen of the nicotinamide ring for stereochemically controlled transfer to the C-3 position of 3-dehydroshikimate. These structural features collectively create a precisely arranged active site environment that facilitates the catalytic reduction of 3-dehydroshikimate to shikimate with high specificity and efficiency, making AroE an excellent target for structure-based inhibitor design targeting the shikimate pathway.

How does the structure of L. monocytogenes AroE compare with that of other bacterial species, and what implications might these differences have for inhibitor design?

The structural characteristics of L. monocytogenes AroE show both conserved features and species-specific variations compared to AroE enzymes from other bacterial pathogens, presenting opportunities for selective inhibitor design. While complete crystallographic data for L. monocytogenes AroE is not explicitly described in the search results, comparative analysis with related bacterial AroE structures can be informative:

Bacterial SpeciesKey Structural DistinctionsImplications for Selective Inhibitor Design
L. monocytogenes vs. E. coliDifferences in substrate binding loop flexibility and surface charge distributionTarget unique binding pocket topography for selectivity
L. monocytogenes vs. M. tuberculosisVariations in NADPH binding site architecture and catalytic residue positioningDesign inhibitors exploiting differential cofactor interactions
L. monocytogenes vs. H. influenzaeDifferences in allosteric regulatory sites and interdomain communicationTarget L. monocytogenes-specific allosteric sites
Gram-positive vs. Gram-negative AroEVariations in surface loops and protein dynamicsExploit differential accessibility and conformational states

These structural distinctions, while subtle, create opportunities for developing inhibitors with enhanced selectivity for L. monocytogenes AroE. The structures of AroE are being used as templates for the synthesis of effective inhibitors of the shikimate pathway , indicating the practical utility of structural information in drug discovery efforts. Species-specific inhibitor design strategies might include: (1) targeting unique substrate binding pocket features using structure-based methods, (2) exploiting differences in protein dynamics and conformational states through molecular dynamics simulations, (3) developing transition-state analogs that leverage subtle differences in catalytic mechanisms, and (4) designing bifunctional inhibitors that simultaneously engage both the substrate and cofactor binding sites, potentially achieving enhanced selectivity through cooperative binding effects.

What methods are most effective for analyzing the kinetic properties of AroE, and how can researchers distinguish between different mechanisms of inhibition?

Analyzing the kinetic properties of AroE and distinguishing between different inhibition mechanisms requires a systematic approach combining multiple complementary techniques. The following methodological framework enables comprehensive kinetic characterization and inhibition mechanism determination:

Analytical MethodExperimental ApproachData InterpretationDistinguishing Features
Steady-state KineticsVary [substrate] at multiple fixed [inhibitor], monitor NADPH absorbance decrease at 340nmLineweaver-Burk, Hanes-Woolf plotsCompetitive: ↑Km, same Vmax
Noncompetitive: Same Km, ↓Vmax
Uncompetitive: ↓Km, ↓Vmax
Mixed: ↑Km, ↓Vmax
Inhibitor Binding StudiesIsothermal Titration Calorimetry (ITC)Binding enthalpy, entropy, and stoichiometryCompetitive inhibitors bind only to free enzyme (E)
Pre-steady-state KineticsStopped-flow spectroscopy with rapid mixingRate constants for individual stepsIdentifies rate-limiting step and mechanism
Structural AnalysisX-ray crystallography of enzyme-inhibitor complexesBinding mode and interaction networksDirectly visualizes inhibitor binding site
Order of Addition StudiesCompare effects of adding inhibitor before/after substrateDifferential inhibition patternsTime-dependent inhibition suggests slow-binding
Temperature DependenceMeasure kinetic parameters at multiple temperaturesArrhenius plots, activation energyMechanism-specific temperature sensitivities

For competitive inhibitors, Dixon plots (1/v versus [I] at different [S]) yield intersecting lines, while for noncompetitive inhibitors, lines converge on the x-axis. Cornish-Bowden plots (S/v versus [I]) provide complementary information - parallel lines for competitive inhibition and intersecting lines for noncompetitive inhibition. For time-dependent inhibitors, progress curves exhibit curvature over time, distinguishing them from rapid-equilibrium inhibitors. Advanced approaches include hydrogen-deuterium exchange mass spectrometry (HDX-MS) to detect inhibitor-induced conformational changes and molecular dynamics simulations to understand inhibitor effects on protein dynamics . These methodologies collectively enable detailed characterization of inhibition mechanisms, providing critical insights for rational optimization of selective AroE inhibitors.

Is there evidence that AroE contributes to the virulence or pathogenicity of Listeria monocytogenes serotype 4b?

While direct experimental evidence specifically connecting AroE to virulence in L. monocytogenes serotype 4b is not explicitly detailed in the search results, multiple lines of indirect evidence suggest its potential contribution to pathogenicity. The shikimate pathway, in which AroE functions as a critical enzyme, is essential for the synthesis of aromatic amino acids and other aromatic compounds that are indispensable for bacterial survival, particularly during infection. Serotype 4b strains belong predominantly to lineage I, which is frequently associated with epidemic human listeriosis outbreaks , suggesting that their metabolic capabilities, potentially including optimized AroE functionality, may contribute to enhanced virulence.

L. monocytogenes must coordinate its metabolic and virulence programs in response to rapidly changing environments within the host to cause disease successfully . This coordination likely includes regulation of essential metabolic pathways like the shikimate pathway. The search results indicate that L. monocytogenes employs sophisticated regulatory mechanisms for adapting to host environments, exemplified by the redox-responsive transcriptional regulator Rex that represses fermentative metabolism and is required for proper virulence gene expression . Similar regulatory mechanisms might modulate AroE activity and the shikimate pathway during infection.

The development of cross-reactive vaccines utilizing common bacterial antigens, including glyceraldehyde-3-phosphate dehydrogenase (GAPDH) from L. monocytogenes, demonstrates the immunological significance of metabolic enzymes in bacterial pathogens . By analogy, AroE likely contributes to the metabolic fitness of L. monocytogenes during infection, potentially supporting growth in nutrient-limited host environments where de novo synthesis of aromatic compounds is essential for survival and pathogenesis.

How does the metabolism of L. monocytogenes, particularly pathways involving AroE, change during infection and host adaptation?

The metabolism of L. monocytogenes undergoes significant rewiring during infection and host adaptation, with pathways involving AroE likely playing crucial roles in this transition. During host invasion and intracellular survival, L. monocytogenes must adapt from environmental saprophytic metabolism to a specialized intracellular metabolic program that enables survival within diverse host microenvironments. The search results indicate that L. monocytogenes employs sophisticated regulatory mechanisms to coordinate its metabolic and virulence programs in response to rapidly changing environments within the host .

The redox-responsive transcriptional regulator Rex represents one such regulatory mechanism, repressing fermentative metabolism and modulating virulence gene expression in response to changes in cellular redox state (NADH/NAD+ ratio) . This dynamic metabolic regulation is essential for L. monocytogenes to transit through the gastrointestinal tract and disseminate to peripheral organs. While specific regulation of the shikimate pathway during infection is not directly addressed in the search results, it is likely subject to similar regulatory control to optimize aromatic compound biosynthesis during infection.

The ability of L. monocytogenes to adapt to diverse host environments, from the acidic conditions of the gastrointestinal tract to the intracellular environment of host cells, requires comprehensive metabolic adaptation. Within host cells, particularly in the nutrient-limited phagosomal environment, biosynthetic pathways including the shikimate pathway would likely be upregulated to compensate for limited nutrient availability. The demonstration that Rex is required for optimal L. monocytogenes growth in the presence of oxygen further underscores the critical interplay between metabolic adaptation and successful pathogenesis, a relationship in which AroE and the shikimate pathway likely play important roles.

Can targeting AroE be an effective strategy for controlling L. monocytogenes infection, and what are the potential advantages compared to targeting other virulence factors?

Targeting AroE presents a promising strategy for controlling L. monocytogenes infection, with several potential advantages compared to traditional virulence factor-based approaches:

AspectTargeting AroE/Shikimate PathwayTargeting Classical Virulence Factors (e.g., LLO)
SelectivityHigh - pathway absent in mammals Variable - some virulence factors have mammalian homologs
EssentialityRequired for bacterial survival regardless of virulence stateMay be dispensable in certain infection stages/conditions
Resistance BarrierHigh - mutations likely compromise fitnessLower - compensatory mechanisms may circumvent inhibition
Activity SpectrumPotentially effective against multiple listeria serotypesOften serotype or strain-specific
Application RangeActive against both replicating and persistent bacteriaOften most effective against actively replicating bacteria
Development ApproachStructure-based design possible with crystallographic data Often requires high-throughput screening approaches

The shikimate pathway's absence in mammals provides an excellent opportunity for selective toxicity, allowing for the development of antimicrobials that target bacterial AroE without affecting host cellular processes . Unlike virulence factors that may be dispensable under certain conditions or subject to strain-specific variations, the shikimate pathway represents an essential metabolic function that cannot be easily circumvented through alternative pathways.

The structures of AroE are being used as templates for the synthesis of effective inhibitors of the shikimate pathway , indicating the feasibility of this approach for drug discovery. Additionally, inhibitors targeting metabolic enzymes like AroE could potentially be effective against both actively replicating bacteria and slow-growing persistent populations, addressing a major challenge in treating chronic infections. While traditional virulence factors like Listeriolysin O (LLO) are important for specific stages of pathogenesis , targeting fundamental metabolic processes offers the potential for broader efficacy across multiple infection stages and bacterial physiological states.

What are the current approaches for designing selective inhibitors of Listeria monocytogenes serotype 4b AroE, and what challenges need to be overcome?

Current approaches for designing selective inhibitors of L. monocytogenes serotype 4b AroE employ integrated strategies combining structural biology, computational methods, and medicinal chemistry. The following table outlines major design approaches and their associated challenges:

Design ApproachMethodological StrategyKey AdvantagesMajor Challenges
Structure-Based DesignUtilize crystal structures for rational inhibitor developmentPrecise targeting of binding pocketsLimited availability of L. monocytogenes-specific AroE structures
Fragment-Based ScreeningIdentify small molecules that bind to different enzyme regionsDiscovers novel chemical scaffoldsFragment linking while maintaining drug-like properties
Transition-State AnaloguesDesign compounds mimicking reaction transition stateHigh-affinity binding potentialComplex synthesis and potential stability issues
Virtual ScreeningComputational evaluation of compound librariesCost-effective initial screeningValidation required through experimental testing
Cofactor-Competitive InhibitorsTarget NADPH binding sitePotentially higher selectivityCompetition with intracellular NADPH concentrations
Allosteric InhibitorsTarget regulatory sites distinct from active siteNovel mechanism of inhibitionIdentifying and characterizing allosteric sites

Additional challenges include developing inhibitors that maintain efficacy against potential resistance mutations without compromising selectivity, optimizing pharmacokinetic properties for appropriate tissue distribution (particularly to the central nervous system for treatment of Listeria meningitis), and identifying synergistic combination approaches with existing antibiotics. Despite these challenges, the fundamental importance of the shikimate pathway and the absence of this pathway in mammals continue to make AroE an attractive target for selective antimicrobial development.

How can CRISPR-Cas9 or other gene editing technologies be utilized to study the function of AroE in L. monocytogenes serotype 4b?

CRISPR-Cas9 and other gene editing technologies offer powerful approaches to investigate AroE function in L. monocytogenes serotype 4b, enabling precise genetic manipulations that were previously challenging to achieve. The following table outlines key applications of these technologies for studying AroE:

Genetic ApproachExperimental StrategyResearch ApplicationsTechnical Considerations
Complete Gene KnockoutCRISPR-Cas9 mediated deletion of aroEEssentiality assessmentMay require conditional approaches if gene is essential
Conditional ExpressionInducible/repressible promoter systemsTemporal control of AroE expressionPotential leaky expression in uninduced state
Point MutationsSingle nucleotide editing of catalytic residuesStructure-function analysisRequires precise sgRNA design and HDR templates
Domain DeletionsTargeted deletion of specific protein domainsDomain function analysisRisk of protein misfolding or instability
Fluorescent TaggingC-terminal fusion with fluorescent proteinsLocalization and expression studiesPotential interference with protein function
CRISPRidCas9-mediated transcriptional repressionTunable gene knockdownVariable repression efficiency across target sites
Allelic ReplacementIntroduction of homologous aroE variantsCross-species complementation studiesRequires efficient homologous recombination

For studying potentially essential genes like aroE, conditional approaches are particularly valuable. These include CRISPRi (CRISPR interference) with a catalytically inactive Cas9 for titratable repression of gene expression, or inducible/repressible promoter systems that allow controlled expression. For investigating AroE's role in pathogenesis, complementation studies can be performed where mutant aroE alleles are introduced at ectopic sites in aroE-deficient strains, followed by virulence assessment in cell culture and animal models.

CRISPR-based approaches can also facilitate the introduction of point mutations in catalytic residues or substrate-binding sites, generating strains with attenuated AroE activity rather than complete loss, enabling the study of partial pathway inhibition. For comprehensive analysis, these genetic approaches should be combined with biochemical assays of shikimate pathway function and virulence assessment in relevant infection models, connecting genotype to phenotype in a physiologically relevant context.

What advanced omics approaches (genomics, transcriptomics, proteomics, metabolomics) can provide insights into the role of AroE in L. monocytogenes biology and pathogenesis?

Advanced omics approaches provide comprehensive, system-level insights into AroE's role in L. monocytogenes biology and pathogenesis by capturing the complex interplay between metabolism and virulence. The following table outlines key omics methodologies and their specific applications for studying AroE:

Omics ApproachExperimental MethodologySpecific Insights for AroE ResearchIntegration with Other Data
Comparative GenomicsWhole genome sequencing of multiple strainsaroE sequence variations across lineages and serotypesCorrelate with virulence phenotypes
TranscriptomicsRNA-seq under various conditionsaroE expression patterns during infection/stressIdentify co-regulated gene clusters
ProteomicsLC-MS/MS with quantitative labeling (TMT/iTRAQ)AroE protein levels and post-translational modificationsConnect to transcriptional regulation
MetabolomicsTargeted LC-MS of shikimate pathway metabolitesFlux through shikimate pathway during infectionLink to downstream aromatic compound synthesis
InteractomicsAffinity purification-mass spectrometryAroE protein-protein interactionsIdentify regulatory partners
Fluxomics13C-labeled substrate tracingCarbon flux through shikimate pathwayQuantify pathway activity
PhenomicsHigh-throughput growth/virulence phenotypingEffects of aroE mutations on multiple phenotypesConnect genotype to phenotype

Comparative genomics across L. monocytogenes strains, particularly contrasting serotype 4b with less virulent serotypes, can reveal polymorphisms in aroE and associated regulatory elements that might contribute to enhanced virulence of epidemic strains. Transcriptomic analyses using RNA-seq under various conditions (different nutrients, stresses, infection stages) can identify co-regulated gene clusters involving aroE, revealing potential regulatory networks connecting metabolism to virulence, similar to the redox-responsive regulation described for other metabolic pathways .

Metabolomics is particularly valuable for studying metabolic enzymes like AroE, allowing quantification of shikimate pathway intermediates and end products under different conditions, directly linking genotype to biochemical phenotype. Integration of multi-omics data through computational approaches can generate testable hypotheses about AroE's broader role in L. monocytogenes' adaptation to various environments encountered during infection. Such integrated analyses may reveal unexpected connections between the shikimate pathway and virulence mechanisms, potentially identifying novel intervention points for controlling L. monocytogenes infections.

What are common challenges in expressing and purifying active recombinant L. monocytogenes AroE, and how can they be addressed?

Researchers commonly encounter several challenges when expressing and purifying active recombinant L. monocytogenes AroE. These issues and their potential solutions are outlined in the following table:

ChallengeCommon SymptomsTroubleshooting ApproachPrevention Strategy
Protein InsolubilityHigh protein in pellet fraction after lysisLower induction temperature (16-20°C)
Reduce IPTG concentration (0.1-0.2 mM)
Use solubility tags (SUMO, MBP)
Optimize codon usage for expression host
Include osmolytes (sorbitol, glycerol) in media
Low Expression YieldMinimal band on SDS-PAGE at expected sizeTest multiple expression strains
Optimize media composition
Extend induction time
Screen multiple construct designs
Use strong T7-based expression systems
Inclusion Body FormationInsoluble aggregates requiring denaturationCo-express chaperones (GroEL/ES)
Use Arctic Express strain
Develop refolding protocol
Express as fusion with highly soluble partners
Include low concentrations of non-ionic detergents
Proteolytic DegradationMultiple bands below expected sizeAdd protease inhibitors
Use protease-deficient strains
Purify at 4°C with reduced processing time
Include stabilizing ligands (NADPH)
Remove flexible regions prone to proteolysis
Activity Loss During PurificationDecreasing specific activity across stepsInclude NADPH (50-100 μM) in buffers
Maintain reducing environment (1-5 mM DTT)
Minimize freeze-thaw cycles
Buffer screening for optimal stability
Add stabilizing agents (glycerol, arginine)
Aggregation During StorageVisible precipitate, increasing turbidityFilter before storage
Store at higher dilution
Test various buffer conditions
Flash-freeze in liquid nitrogen
Add 10-20% glycerol before freezing

Activity loss during purification often results from oxidation of catalytic cysteines or cofactor dissociation. This can be mitigated by including reducing agents (DTT, TCEP) and cofactors (NADPH) in purification buffers . For long-term storage, flash-freezing small aliquots in liquid nitrogen with 10-20% glycerol and storage at -80°C typically preserves enzymatic activity, minimizing the detrimental effects of repeated freeze-thaw cycles.

How can researchers troubleshoot inconsistent results in AroE activity assays and ensure reliable kinetic measurements?

Troubleshooting inconsistent results in AroE activity assays requires systematic investigation of multiple factors that can affect enzyme kinetics and measurement reliability. The following table outlines common sources of variability and their solutions:

Source of VariabilityDiagnostic IndicatorsTroubleshooting ApproachQuality Control Measures
Buffer Composition VariationsInconsistent baseline activityStandardize buffer preparation
Test pH and ionic strength effects
Evaluate buffer component interference
Prepare master buffer stocks
Document exact composition
Check pH before each experiment
NADPH Quality/StabilityDecreasing blank absorbance
Non-linear standard curves
Prepare fresh NADPH solutions
Protect from light
Store concentrated stocks at -80°C
Include NADPH-only controls
Establish acceptance criteria
Determine NADPH stability curve
Temperature FluctuationsDrift in reaction rates
Inconsistent Km/Vmax values
Pre-equilibrate all components
Use temperature-controlled cuvette holders
Monitor actual temperature in reaction vessel
Perform assays at fixed temperature
Include internal standards
Document temperature throughout experiment
Enzyme Concentration IssuesNon-linear enzyme-rate relationship
Poor reproducibility
Determine linear range for enzyme concentration
Use consistent protein quantification method
Adjust for batch-to-batch variations
Express results as specific activity
Include enzyme dilution series
Verify enzymatic purity by SDS-PAGE
Substrate Stability/PurityDecreasing activity over time
Variable kinetic parameters
Test substrate stability under assay conditions
Verify substrate purity
Prepare fresh substrate solutions
Store substrates as concentrated aliquots
Document lot-to-lot variations
Include substrate stability controls
Spectrophotometer LimitationsSignal drift
Poor baseline stability
Regular instrument calibration
Background correction at reference wavelength
Minimize environmental light interference
Establish minimum absorbance change criteria
Use appropriate blanks
Perform regular instrument performance checks

For particularly challenging kinetic analyses, alternative assay methods can provide complementary data to validate spectrophotometric results. These include coupled enzyme assays (linking AroE activity to a more easily measured secondary reaction), HPLC-based product quantification for direct measurement of shikimate formation, or isothermal titration calorimetry for thermodynamic characterization of the reaction.

Statistical robustness should be ensured through multiple independent experiments with technical replicates and appropriate control reactions. Data analysis should include careful evaluation of linear reaction ranges, application of appropriate kinetic models, and statistical testing to identify significant differences. Following these rigorous approaches will significantly improve the reliability and reproducibility of AroE activity measurements across different experimental conditions and laboratory settings.

What strategies can help overcome the challenges of studying AroE function in the context of intracellular infection models?

Studying AroE function during intracellular infection presents unique challenges requiring specialized approaches that integrate bacterial genetics, cell biology, and analytical biochemistry. The following table outlines strategies to address these challenges:

ChallengeExperimental StrategyTechnical ApproachAnalytical Considerations
Distinguishing Bacterial vs. Host MetabolismSelective Labeling Approaches13C-labeled precursors specific to shikimate pathway
Bacterial-specific promoters driving reporter constructs
MS-based metabolite identification
Signal normalization to bacterial burden
Background subtraction from uninfected controls
Temporal Regulation of AroEConditional Expression SystemsTetracycline-inducible promoters
Destabilization domains for protein-level control
CRISPRi for titratable repression
Western blotting for expression verification
qRT-PCR for transcript analysis
Time-course metabolite analysis
Spatial Heterogeneity in InfectionSingle-Cell AnalysisFluorescent reporters linked to aroE expression
Micro-dissection of infected tissues
Laser capture microdissection
Flow cytometry for population analysis
Confocal microscopy for spatial distribution
Single-cell transcriptomics
Low Bacterial Numbers in InfectionSensitive Detection MethodsDigital PCR for absolute quantification
Nanostring for direct mRNA counting
Highly selective MRM-MS for metabolites
Careful selection of normalization strategies
Standard curves with known concentrations
Statistical approaches for low-abundance data
Multiple Bacterial SubpopulationsPopulation Separation TechniquesFACS sorting of differentially labeled bacteria
Density gradient separation
Single-cell encapsulation
Analysis of population heterogeneity
Identification of distinct metabolic states
Correlation with virulence characteristics

For tracking AroE activity during infection, fluorescent biosensors responsive to shikimate pathway metabolites can provide real-time readouts in living infected cells. Advanced microscopy techniques, including super-resolution and correlative light-electron microscopy, can identify the subcellular localization of AroE and associated metabolic enzymes during infection.

Chemical-genetic approaches using partial inhibition of AroE with sublethal inhibitor concentrations can complement genetic studies while avoiding the complications of complete gene deletion if aroE is essential. Development of cell-type specific infection models (hepatocytes, macrophages, enterocytes) can reveal how AroE's importance might vary in different host cell environments, reflecting the tissue tropism observed in L. monocytogenes infections. These approaches collectively enable a more comprehensive understanding of AroE function in the complex environment of intracellular infection, potentially revealing new aspects of host-pathogen metabolic interactions.

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.