Recombinant Listeria monocytogenes serotype 4b Glucosamine-6-phosphate deaminase (nagB)

Shipped with Ice Packs
In Stock

Description

Enzymatic Function and Physiological Role

Glucosamine-6-phosphate deaminase (NagB) is a cytoplasmic enzyme encoded by the nagB gene (lmo0957 in Listeria monocytogenes). It catalyzes the reversible conversion of glucosamine-6-phosphate (GlcN-6-P) to fructose-6-phosphate (F6P) and ammonia, a critical step in amino sugar metabolism and peptidoglycan recycling . In L. monocytogenes serotype 4b, NagB is integral to:

  • Cell wall biosynthesis: GlcN-6-P serves as a precursor for UDP-N-acetylglucosamine (UDP-GlcNAc), essential for peptidoglycan and lipopolysaccharide synthesis .

  • Stress adaptation: NagB is upregulated under high-pressure processing (HPP) stress, aiding bacterial survival by enhancing peptidoglycan recycling .

  • Carbon metabolism: Links amino sugar catabolism to glycolysis via F6P production, supporting energy generation during nutrient limitation .

Recombinant Expression and Applications

Recombinant NagB is produced by cloning the nagB gene into heterologous systems (e.g., E. coli) for functional studies. Key findings from recombinant studies include:

Table 1: Phenotypic Effects of nagB Mutations in L. monocytogenes

ParameterWild-TypeΔnagB MutantSource
Peptidoglycan contentNormalReduced by 40–60%
Stress resistanceHighSensitive to mutanolysin, colistin
GlcNAc productionLowIncreased by 84% (in L. plantarum ΔnagB)

Table 2: Kinetic Properties of NagB

SubstrateKm (mM)Vmax (μmol/min/mg)Allosteric Activator
GlcN-6-P0.8–1.212.4 ± 1.3GlcNAc-6-P
F6P (reverse reaction)3.5–4.18.9 ± 0.7None

Role in Virulence and Pathogenesis

NagB contributes to L. monocytogenes pathogenicity through:

  • Biofilm formation: GlcN-6-P depletion in ΔnagB mutants reduces biofilm biomass by 30–50% under osmotic stress .

  • Intracellular survival: σB-mediated upregulation of nagB enhances bacterial resilience in host macrophages .

  • Cross-protective immunity: Recombinant NagB shares epitopes with homologous enzymes in Mycobacterium and Streptococcus, suggesting potential for broad-spectrum vaccine development .

Industrial and Biotechnological Relevance

Recombinant NagB has been engineered for bioproduction applications:

  • GlcNAc synthesis: Inactivation of nagB in Lactobacillus plantarum increased GlcNAc yield to 797 mg/L .

  • Metabolic engineering: Fusion of NagB with glmS (glutamine-fructose-6-phosphate aminotransferase) optimized carbon flux in synthetic pathways .

Research Gaps and Future Directions

  • Structural dynamics: The allosteric activation mechanism by GlcNAc-6-P remains unresolved in L. monocytogenes .

  • Therapeutic potential: Recombinant NagB’s cross-reactive epitopes warrant exploration in anti-infective vaccines .

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 consolidate 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% and can serve as a 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 maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
nagB; LMOf2365_0977; Glucosamine-6-phosphate deaminase; EC 3.5.99.6; GlcN6P deaminase; GNPDA; Glucosamine-6-phosphate isomerase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-234
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
nagB
Target Protein Sequence
MQLITTENKL AGSKKALEII EKGITSGEVN TLGLATGSTP ETLYAELVKS DVDTKNVTTT NLDEYVGLAA NDPNSYHYYM NELLFSKKAF KESFLPNGEA TDAEAECARY EEILSEHPID IQVLGIGTNG HIGFNEPGTP FDSLTHKVVL TDSTREANKR FFEREEDVPT HAYSMGIKSI MNAKKIILLA FGENKAQAIK ETIKGPVDVN CPASVLQNHP DVTVILDNEA ASLL
Uniprot No.

Target Background

Function

Function: Catalyzes the reversible isomerization-deamination of glucosamine 6-phosphate (GlcN6P) to fructose 6-phosphate (Fru6P) and ammonium ion.

Database Links
Protein Families
Glucosamine/galactosamine-6-phosphate isomerase family, NagB subfamily

Q&A

What is the functional significance of nagB in Listeria monocytogenes serotype 4b?

Glucosamine-6-phosphate deaminase (nagB) plays a critical role in amino sugar metabolism within Listeria monocytogenes serotype 4b. This enzyme catalyzes the conversion of glucosamine-6-phosphate (GlcN-6P) to fructose-6-phosphate (Fru-6P) and ammonia, representing the final step in the amino sugar-specific catabolic pathway. This conversion is essential for redirecting amino sugars into the glycolytic pathway, making nagB a crucial enzyme for bacterial metabolism and growth. The enzyme serves as a key junction between nitrogen metabolism and carbon utilization pathways, with significant implications for cellular energy production. In Listeria monocytogenes serotype 4b, this function is particularly important as this serotype is responsible for a substantial fraction of food-borne listeriosis cases and is involved in the majority of common-source outbreaks .

How is nagB regulated in Listeria monocytogenes?

NagB in Listeria monocytogenes, similar to its homolog in E. coli, is subject to complex allosteric regulation through multiple mechanisms. The enzyme is allosterically activated by N-acetylglucosamine 6-phosphate (GlcNAc-6P), which acts as a metabolic signal of amino sugar availability. Additionally, nagB is regulated through direct protein-protein interactions with at least three regulatory proteins: the phosphocarrier protein HPr, N-acetylmannosamine-6-phosphate 2-epimerase (NanE), and the uridylylated form of the nitrogen regulatory PII protein (U-PII) .

These regulators respond to different physiological stimuli:

  • HPr activation requires the presence of GlcNAc-6P and reflects the availability of extracellular PTS sugar substrates

  • NanE activates nagB regardless of GlcNAc-6P presence and is essential for neuraminic acid and N-acetylmannosamine utilization

  • U-PII (but not unmodified PII) activates nagB more than 10-fold at low substrate concentrations, but only in the presence of GlcNAc-6P

These complex regulatory mechanisms allow Listeria to fine-tune nagB activity in response to carbon and nitrogen availability, adapting metabolism to different growth conditions.

What techniques are commonly used to identify Listeria monocytogenes serotype 4b?

Identification of Listeria monocytogenes serotype 4b typically employs a combination of traditional microbiological methods and modern molecular techniques. Researchers have developed "molecular serotyping" approaches that utilize serotype-specific sequences unique to major serotypes of epidemiological relevance, particularly serotype 4b. The methodological approach involves:

  • Identification of candidate serotype-specific sequences unique to serotype 4b

  • Design of oligonucleotide primers based on these sequences

  • Amplification of corresponding DNA fragments using Polymerase Chain Reaction (PCR)

  • Validation of amplified fragments as probes against genomic DNAs from diverse strain panels

  • Combination with DNA subtyping tools derived from flagellin gene sequences to enhance resolution

Additionally, Southern blot techniques using digoxigenin-labeled probes derived from serotype-specific genes can more accurately determine the distribution of these sequences in different L. monocytogenes strains. This approach enables precise identification of serotype 4b isolates from both clinical and food samples, supporting epidemiological investigations and research studies .

What are the recommended protocols for cloning and expressing recombinant nagB from Listeria monocytogenes serotype 4b?

For successful cloning and expression of recombinant nagB from L. monocytogenes serotype 4b, researchers should follow this methodological workflow:

  • Primer Design: Design primers targeting the nagB gene with appropriate restriction sites for directional cloning. Include a 5' overhang for efficient enzyme digestion and incorporate a His-tag or other purification tag if needed.

  • PCR Amplification: Extract genomic DNA from L. monocytogenes serotype 4b using standardized protocols. Amplify the nagB gene using high-fidelity polymerase to minimize introduction of mutations. Optimize PCR conditions through gradient PCR to determine ideal annealing temperatures.

  • Expression Vector Selection: Select an appropriate expression vector based on research needs. For structural studies, vectors with T7 promoters in E. coli BL21(DE3) systems work well. For functional studies comparing wild-type and mutant forms, consider inducible systems with tight regulation.

  • Transformation and Expression: Transform the recombinant plasmid into an appropriate host (typically E. coli). Optimize expression conditions by testing various induction parameters:

    • Temperature range: 16-37°C

    • IPTG concentration: 0.1-1.0 mM

    • Induction time: 4-24 hours

  • Purification Strategy: Implement a two-step purification approach:

    • Initial affinity chromatography (Ni-NTA for His-tagged protein)

    • Size exclusion chromatography for higher purity

Monitor protein quality with SDS-PAGE and Western blotting at each stage of purification. Confirm enzymatic activity using a spectrophotometric assay measuring the conversion of GlcN-6P to Fru-6P through coupled enzyme reactions .

How can researchers effectively measure allosteric regulation of nagB activity?

Measuring the allosteric regulation of nagB requires careful experimental design to quantify enzymatic activity under various conditions. A comprehensive approach includes:

  • Baseline Activity Determination: Measure nagB activity using a coupled enzyme assay system where the production of Fru-6P is linked to NADH oxidation via phosphoglucose isomerase and glucose-6-phosphate dehydrogenase. Monitor the decrease in absorbance at 340 nm spectrophotometrically.

  • Allosteric Activator Titration: Perform enzyme assays with increasing concentrations of GlcNAc-6P (0-5 mM) to establish dose-response curves. Calculate kinetic parameters (Km and Vmax) at each concentration to determine the degree of allosteric activation.

  • Protein Activator Analysis: To investigate protein-protein interactions affecting nagB activity:

    • Express and purify regulatory proteins (HPr, NanE, and PII)

    • For PII protein, prepare both unmodified and uridylylated forms

    • Test each protein individually at various concentrations (typically 0.1-10 μM)

    • Examine combinations of multiple regulators to detect synergistic effects

  • Data Analysis: Use enzyme kinetics software to fit data to appropriate allosteric models (e.g., Hill equation, Monod-Wyman-Changeux model). Calculate activation constants and cooperativity coefficients.

Research has demonstrated that uridylylated PII activates nagB >10-fold at low substrate concentrations, NanE increases activity >2-fold, and combinations of activators can produce synergistic effects. For example, HPr and U-PII activation requires GlcNAc-6P, while NanE can activate nagB regardless of GlcNAc-6P presence .

What experimental approaches can be used to investigate the role of nagB in Listeria monocytogenes virulence?

Investigating the role of nagB in L. monocytogenes virulence requires a multi-faceted approach combining molecular genetics, cell culture, and animal models:

  • Gene Knockout and Complementation:

    • Create a nagB deletion mutant using allelic exchange techniques

    • Develop complemented strains expressing wild-type nagB

    • Generate point mutants affecting catalytic activity or regulatory interactions

    • Confirm genotypes by PCR and sequencing; verify phenotypes by growth curves in various carbon sources

  • Cell Culture Infection Models:

    • Human umbilical vein endothelial cells (HUVEC) and brain microvascular endothelial cells (BMEC) provide relevant models

    • Quantify bacterial adhesion, invasion, and intracellular replication

    • Measure host cell responses through ELISA-based detection of activation markers (E-selectin, ICAM-1, VCAM-1)

    • Compare wild-type, ΔnagB mutant, and complemented strains

  • Transcriptional Analysis:

    • Perform RNA-Seq or qRT-PCR to identify genes differentially expressed in ΔnagB mutants

    • Focus on virulence genes and metabolic pathways affected by nagB deletion

    • Compare expression patterns under various growth conditions mimicking host environments

  • In vivo Infection Models:

    • Mouse listeriosis model to evaluate bacterial dissemination to organs

    • Measure bacterial loads in liver, spleen, and brain

    • Assess survival curves and histopathological changes

This comprehensive approach enables researchers to determine whether nagB contributes to virulence directly through interactions with host factors or indirectly by affecting bacterial metabolism and stress responses during infection .

How do the regulatory mechanisms of nagB differ between Listeria monocytogenes and other bacterial species?

The regulatory mechanisms governing nagB activity show interesting differences between Listeria monocytogenes and other bacterial species, particularly Escherichia coli where nagB has been extensively studied:

  • Allosteric Regulation:

    • Both L. monocytogenes and E. coli nagB are allosterically activated by GlcNAc-6P

    • E. coli nagB shows a stronger response to GlcNAc-6P with a dissociation constant (K) of 2.1 mM

    • L. monocytogenes nagB may have evolved different binding affinities reflecting its pathogenic lifestyle

  • Protein-Protein Interactions:

    • Both species demonstrate regulation through multiple protein partners

    • E. coli nagB is regulated by HPr, NanE, and uridylylated PII protein

    • Differences exist in the magnitude of activation: U-PII activates E. coli nagB >10-fold at low substrate concentrations

  • Transcriptional Control:

    • E. coli nagB is encoded in an operon with nagA and regulated by NagC transcriptional regulator

    • E. coli nagB expression is induced only when amino sugars are present in the medium

    • L. monocytogenes may integrate signals from both nutrient availability and virulence regulatory networks

  • Metabolic Integration:

    • In E. coli, nagB directs GlcN-6P to the glycolytic pathway, opposing the anabolic function of GlmS in peptidoglycan synthesis

    • In L. monocytogenes, this metabolic junction may serve additional functions related to virulence

The table below summarizes key proteins that interact with nagB based on protein interaction studies:

ProteinCellular LocationInteraction ScoreProtein Function and EC Number
NanECytoplasm5.4N-Acetylmannosamine-6-phosphate 2-epimerase (EC 5.1.3.9)
GlnBCytoplasm5.3Nitrogen regulatory protein P-II
PtsHMembrane-associated5.3Phosphocarrier protein HPr

These differences reflect the adaptation of nagB regulation to specific ecological niches and metabolic requirements of each bacterial species .

What approaches are recommended for resolving contradictory data in nagB functional studies?

When faced with contradictory data in nagB functional studies, researchers should implement a systematic troubleshooting approach:

  • Standardize Experimental Conditions:

    • Ensure consistent buffer compositions, pH, and temperature across all experiments

    • Standardize protein purification protocols to minimize variability in enzyme preparations

    • Verify enzyme stability under storage and assay conditions through time-course activity measurements

  • Validate Protein Quality:

    • Perform circular dichroism spectroscopy to confirm proper protein folding

    • Use size exclusion chromatography to detect aggregation or oligomerization states

    • Implement thermal shift assays to assess protein stability under various conditions

  • Cross-Validate Activity Assays:

    • Compare results from multiple independent assay methods:

      • Direct measurement of ammonia production

      • Coupled enzymatic assays tracking Fru-6P formation

      • Isothermal titration calorimetry for binding studies

  • Genetic Validation:

    • Generate clean deletion mutants and complemented strains

    • Perform site-directed mutagenesis targeting specific residues

    • Validate phenotypes across multiple genetic backgrounds

  • Address Strain Variability:

    • Test multiple isolates of L. monocytogenes serotype 4b

    • Sequence the nagB gene and regulatory regions from strains showing discrepant results

    • Consider genomic context and potential polar effects in genetic manipulations

  • Computational Analysis:

    • Use molecular modeling to predict structural impacts of experimental conditions

    • Perform network analysis to identify potential compensatory pathways

    • Apply statistical methods appropriate for the specific data type to determine significance

By systematically addressing these factors, researchers can resolve contradictions and develop a more accurate understanding of nagB function in L. monocytogenes serotype 4b.

What are the optimal conditions for assaying nagB enzyme activity in vitro?

For reliable and reproducible measurement of nagB activity in vitro, researchers should consider these optimized methodological parameters:

  • Buffer Composition and pH:

    • Recommended buffer: 50 mM Tris-HCl at pH 7.5-8.0

    • Include stabilizing agents: 5 mM MgCl₂ and 1 mM DTT

    • Control ionic strength with 100 mM KCl

  • Temperature and Reaction Time:

    • Optimal temperature range: 30-37°C (balance between activity and stability)

    • Linear reaction phase typically maintained for 5-10 minutes

    • Pre-incubate components at assay temperature before initiating reaction

  • Substrate Preparation:

    • Use freshly prepared GlcN-6P solutions

    • Standard concentration range: 0.1-5.0 mM for kinetic studies

    • Include substrate controls to account for non-enzymatic degradation

  • Detection Methods:

    • Direct method: Measure ammonia production using Berthelot's reaction (colorimetric)

    • Coupled method: Link Fru-6P production to NADH oxidation via auxiliary enzymes

    • Recommended coupling enzymes: phosphoglucose isomerase and glucose-6-phosphate dehydrogenase

  • Activator Inclusions:

    • When studying regulation, include GlcNAc-6P at 0.5-2.0 mM

    • For protein activators, maintain molar ratios of activator:nagB between 1:1 and 5:1

    • Remember that U-PII activation requires GlcNAc-6P co-presence, while NanE activation does not

  • Data Collection:

    • For spectrophotometric assays, measure at 340 nm (NADH absorbance)

    • Take readings every 15-30 seconds for accurate initial velocity determination

    • Run reactions in triplicate with appropriate blanks and controls

These optimized conditions allow for precise measurement of nagB activity across various experimental scenarios, enabling reliable comparison between wild-type and mutant forms or between different regulatory states.

How can researchers effectively model the allosteric interactions between nagB and its regulatory proteins?

Modeling the complex allosteric interactions between nagB and its regulatory proteins requires an integrated computational and experimental approach:

  • Structural Analysis:

    • Obtain crystal structures of nagB alone and in complex with regulatory proteins

    • When crystal structures aren't available, use homology modeling based on related proteins

    • Perform molecular dynamics simulations to understand conformational changes upon binding

  • Docking Studies:

    • Use computational docking to predict protein-protein interaction interfaces

    • Validate predictions through mutagenesis of key residues at predicted interfaces

    • Implement ensemble docking to account for protein flexibility

  • Kinetic Modeling:

    • Develop mathematical models incorporating multiple allosteric effectors

    • Consider sequential or simultaneous binding scenarios

    • Use experimental data to constrain model parameters

  • Network Analysis:

    • Map the complete interaction network of nagB and its regulators

    • Identify potential synergistic or antagonistic relationships

    • Research has shown that activation of nagB by HPr and uridylylated PII, as well as by NanE and HPr (but not by NanE and U-PII), is synergistic

  • Experimental Validation:

    • Design mutants targeting specific interaction interfaces

    • Perform binding studies using techniques like surface plasmon resonance

    • Measure activity changes with various combinations of regulators

  • Integration of Multiple Data Types:

    • Combine structural, kinetic, and cellular data into comprehensive models

    • Use systems biology approaches to understand contextual regulation

    • Apply machine learning to predict responses to novel conditions

This integrated approach provides insights into how multiple regulatory inputs are processed by nagB to fine-tune its activity in response to changing metabolic conditions, particularly at the intersection of carbon and nitrogen metabolism .

What are the challenges and solutions in producing site-directed mutants of nagB to study its catalytic mechanism?

Creating and characterizing site-directed mutants of nagB presents several challenges, but implementing specific solutions can yield valuable insights into the enzyme's catalytic mechanism:

  • Challenges in Mutant Design:

    • Identifying catalytically important residues without disrupting protein folding

    • Distinguishing between residues involved in catalysis versus substrate binding

    • Avoiding disruption of regulatory protein interaction sites

    Solutions:

    • Perform sequence alignment across diverse bacterial species to identify conserved residues

    • Use structural information to guide mutation selection

    • Consider conservative substitutions that maintain similar physicochemical properties

  • Expression Difficulties:

    • Reduced stability of mutant proteins leading to aggregation

    • Altered folding kinetics affecting yield

    • Potential toxicity to expression hosts

    Solutions:

    • Lower induction temperature (16-20°C) to slow protein synthesis and improve folding

    • Co-express with molecular chaperones (GroEL/GroES)

    • Use specialized E. coli strains designed for difficult protein expression

  • Activity Assessment Challenges:

    • Distinguishing complete loss of activity from reduced activity

    • Accounting for changes in protein stability affecting apparent activity

    • Detecting subtle changes in regulatory responses

    Solutions:

    • Develop highly sensitive assays capable of detecting low-level activity

    • Normalize activity to actual protein concentration determined by quantitative methods

    • Include thermal stability assays alongside activity measurements

  • Regulatory Interaction Analysis:

    • Mutations may have different effects on different regulatory mechanisms

    • Complex interplay between substrate binding, catalysis, and regulation

    Solutions:

    • Systematically test mutants with each regulatory protein individually and in combination

    • Create mutant panels targeting different functional domains

    • Compare kinetic parameters under various regulatory conditions

  • Structural Verification:

    • Confirming that mutations do not cause major structural changes

    • Validating the specific local changes intended by the mutation

    Solutions:

    • Perform circular dichroism to confirm secondary structure maintenance

    • Use differential scanning fluorimetry to assess thermal stability

    • When possible, solve crystal structures of key mutants

By addressing these challenges systematically, researchers can generate a comprehensive understanding of the structure-function relationships in nagB and how they relate to its dual role in metabolism and potential contributions to virulence in L. monocytogenes serotype 4b.

How can nagB be utilized as a potential target for antimicrobial development against Listeria monocytogenes?

Developing antimicrobials targeting nagB requires a multidisciplinary approach that leverages its essential metabolic function in Listeria monocytogenes:

  • Target Validation:

    • Confirm essentiality through conditional knockdown systems in various growth conditions

    • Demonstrate growth inhibition when nagB function is compromised

    • Verify attenuation of virulence in infection models when nagB is inhibited

  • Inhibitor Design Strategies:

    • Structure-based drug design using crystal structures of L. monocytogenes nagB

    • Focus on three potential binding sites:

      • Active site (competitive inhibitors)

      • Allosteric sites (non-competitive inhibitors)

      • Protein-protein interaction interfaces (disrupting regulatory networks)

    • Virtual screening against compound libraries to identify initial hits

  • Selectivity Considerations:

    • Compare structural differences between bacterial and human homologs

    • Target Listeria-specific regulatory mechanisms

    • Design inhibitors exploiting unique structural features of the serotype 4b variant

  • Efficacy Testing Methodology:

    • Enzymatic assays to confirm direct inhibition

    • Bacterial growth assays in various media compositions

    • Cell infection models to assess impact on virulence

    • Evaluation in animal models of listeriosis

  • Resistance Development Assessment:

    • Perform long-term passage experiments with sub-inhibitory concentrations

    • Sequence nagB and regulatory elements in resistant isolates

    • Model potential resistance mechanisms through structural analysis

  • Delivery Considerations:

    • Evaluate cell permeability of candidate inhibitors

    • Consider prodrug approaches if necessary

    • Test combination approaches with existing antibiotics

This systematic approach harnesses the critical metabolic role of nagB while addressing the challenges inherent in antimicrobial development against an intracellular pathogen like L. monocytogenes serotype 4b.

What are the methodological approaches for studying the role of nagB in Listeria monocytogenes biofilm formation?

Investigating the role of nagB in L. monocytogenes biofilm formation requires specialized techniques spanning molecular genetics, microscopy, and biochemical analysis:

  • Genetic Manipulation Approach:

    • Generate nagB deletion mutants, complemented strains, and regulatory mutants

    • Create fluorescent protein fusions for real-time monitoring

    • Develop inducible expression systems to modulate nagB levels during biofilm development

  • Biofilm Formation Assays:

    • Static microtiter plate assay with crystal violet staining for quantification

    • Flow cell systems to mimic dynamic environments

    • Surface materials relevant to food processing environments:

      • Stainless steel

      • Glass

      • Food-contact polymers

    • Compare biofilm formation under various nutrient conditions, particularly varying amino sugar availability

  • Microscopic Analysis:

    • Confocal laser scanning microscopy to determine:

      • Biofilm architecture

      • Spatial distribution of cells

      • Extracellular matrix composition

    • Electron microscopy for high-resolution structural details

    • Live/dead staining to assess viability within biofilms

  • Biochemical Characterization:

    • Analyze extracellular polymeric substance (EPS) composition:

      • Polysaccharides

      • Proteins

      • Extracellular DNA

    • Measure metabolic activity within biofilms using fluorescent indicators

    • Quantify amino sugar metabolism within biofilm communities

  • Transcriptional Analysis:

    • RNA-Seq or qRT-PCR to compare expression profiles:

      • Planktonic vs. biofilm cells

      • Wild-type vs. nagB mutants

      • Various stages of biofilm development

    • Focus on connections between amino sugar metabolism and known biofilm regulators

  • Stress Response Correlation:

    • Test biofilm resistance to antimicrobials

    • Evaluate persistence under nutrient limitation

    • Assess survival under disinfectant challenge

This methodological framework allows researchers to comprehensively evaluate how nagB and its regulation of amino sugar metabolism influences biofilm formation, potentially revealing new targets for biofilm control in food safety applications.

How can isotope labeling techniques be applied to track nagB-mediated metabolic flux in Listeria monocytogenes?

Isotope labeling provides powerful tools for tracing metabolic pathways involving nagB in Listeria monocytogenes. The following methodological approach outlines how to implement these techniques effectively:

  • Isotope Selection and Labeling Strategies:

    • ¹³C-labeled glucose or glucosamine as primary carbon sources

    • ¹⁵N-labeled amino sugars to track nitrogen transfer

    • Design labeling patterns to distinguish between:

      • Direct flux through nagB pathway

      • Alternative metabolic routes

      • Recycling of metabolic intermediates

  • Experimental Setup:

    • Cultivate L. monocytogenes in chemically defined media with labeled substrates

    • Compare wild-type, nagB mutant, and complemented strains

    • Sample at multiple time points to capture dynamic metabolic changes

    • Include relevant physiological conditions (e.g., low glucose, high amino sugars)

  • Analytical Methods:

    • Gas Chromatography-Mass Spectrometry (GC-MS) for primary metabolite analysis

    • Liquid Chromatography-Mass Spectrometry (LC-MS) for phosphorylated intermediates

    • Nuclear Magnetic Resonance (NMR) spectroscopy for detailed positional isotope analysis

    • High-resolution MS to determine exact isotope incorporation patterns

  • Data Analysis Approaches:

    • Calculate Mass Isotopomer Distribution (MID) for key metabolites

    • Implement flux balance analysis with isotope constraints

    • Develop computational models of central carbon metabolism including the nagB pathway

  • Specific Metabolic Questions to Address:

    • Quantify the contribution of nagB to carbon flux into glycolysis

    • Determine the fate of nitrogen released by nagB deamination

    • Assess how regulatory proteins (HPr, NanE, U-PII) affect metabolic flux distribution

    • Measure how nutrient conditions alter flux through nagB vs. alternative pathways

  • Integration with Transcriptomics/Proteomics:

    • Correlate metabolic flux with gene expression patterns

    • Identify regulatory responses to metabolic perturbations

    • Build comprehensive models of metabolic adaptation

This approach provides quantitative insights into how nagB functions within the broader metabolic network of L. monocytogenes, revealing its importance in nutrient utilization and potentially identifying metabolic vulnerabilities that could be exploited for targeted interventions.

How might nagB function differently across various Listeria monocytogenes serotypes and what methodologies are best for comparative studies?

Investigating serotype-specific differences in nagB function requires systematic comparative approaches across multiple dimensions:

  • Sequence and Structure Analysis:

    • Perform comprehensive sequence alignment of nagB across all L. monocytogenes serotypes

    • Identify serotype-specific amino acid substitutions

    • Generate homology models to predict structural consequences of variations

    • Focus particularly on differences between serotype 4b (highly associated with outbreaks) and other serotypes

  • Expression Pattern Comparison:

    • Quantify nagB expression levels across serotypes using RT-qPCR

    • Determine if regulatory mechanisms differ between serotypes

    • Map promoter regions and transcription factor binding sites

    • Assess expression under various environmental conditions relevant to food contamination and host infection

  • Enzymatic Activity Characterization:

    • Purify recombinant nagB from multiple serotypes

    • Compare kinetic parameters (Km, Vmax, kcat)

    • Assess sensitivity to allosteric regulators

    • Evaluate protein-protein interactions with regulatory partners

  • Genetic Complementation Studies:

    • Create a standardized nagB deletion in a reference strain

    • Complement with nagB variants from different serotypes

    • Evaluate phenotypic restoration in:

      • Growth on amino sugar substrates

      • Stress resistance

      • Virulence characteristics

  • Metabolomic Profiling:

    • Compare metabolite profiles between serotypes when grown on amino sugars

    • Identify differential accumulation of pathway intermediates

    • Use isotope labeling to track carbon flow

    • Correlate metabolic differences with nagB sequence variations

  • Host-Pathogen Interaction Assessment:

    • Compare serotypes in cell invasion assays

    • Evaluate intracellular replication efficiency

    • Assess activation of host immune responses

    • Determine if nagB contributes differently to virulence across serotypes

This comprehensive comparative approach can reveal whether serotype-specific variations in nagB contribute to the epidemiological differences observed between L. monocytogenes serotypes, particularly the prevalence of serotype 4b in human listeriosis outbreaks .

What are the current techniques for exploring the potential cross-talk between nagB and virulence gene expression in Listeria monocytogenes?

Investigating the relationship between metabolic enzymes like nagB and virulence gene expression requires sophisticated approaches that bridge metabolism and pathogenicity:

  • Transcriptome-Wide Association Studies:

    • Compare global transcriptional profiles between:

      • Wild-type and nagB mutant strains

      • Various growth conditions altering nagB activity

    • Implement RNA-Seq with differential expression analysis

    • Focus on known virulence genes regulated by PrfA, the master virulence regulator

  • Metabolite-Mediated Regulation Analysis:

    • Test if metabolic intermediates linked to nagB activity affect virulence gene expression

    • Measure intracellular concentrations of key metabolites (GlcN-6P, Fru-6P, GlcNAc-6P)

    • Correlate metabolite levels with virulence gene expression

    • Test direct addition of cell-permeable analogs of these metabolites

  • Protein-Protein Interaction Networks:

    • Perform pull-down assays with tagged nagB to identify interaction partners

    • Use bacterial two-hybrid systems to screen for interactions with regulators

    • Implement proximity labeling approaches (BioID, APEX) to capture transient interactions

    • Specifically test for interactions with virulence regulators

  • Chromatin Immunoprecipitation Techniques:

    • Determine if metabolic regulators affected by nagB bind to virulence gene promoters

    • Implement ChIP-Seq to map genome-wide binding patterns

    • Look for enrichment of binding sites near virulence genes

    • Test how metabolic conditions alter binding patterns

  • Reporter System Approaches:

    • Create fluorescent reporter fusions to key virulence genes

    • Monitor expression in real-time during metabolic shifts

    • Test specific effects of nagB deletion or overexpression

    • Implement cell-sorting to isolate and characterize subpopulations

  • Host-Relevant Condition Testing:

    • Examine cross-talk under conditions mimicking the host environment:

      • Glucose-limited conditions

      • Presence of host-derived amino sugars

      • Intracellular-like ion concentrations

    • Use cell infection models to validate findings in a relevant context

This integrated approach can reveal how L. monocytogenes coordinates its metabolic state with virulence gene expression, potentially identifying new regulatory mechanisms that contribute to its success as a pathogen, particularly in serotype 4b strains associated with severe outbreaks .

How can structural biology approaches advance our understanding of nagB function and regulation in Listeria monocytogenes?

Advanced structural biology methodologies offer powerful tools for elucidating the molecular mechanisms underlying nagB function and regulation in Listeria monocytogenes:

  • High-Resolution Structure Determination:

    • X-ray crystallography of:

      • Apo-enzyme

      • Substrate-bound complex

      • Enzyme-activator complexes (with GlcNAc-6P, HPr, NanE, U-PII)

    • Cryo-electron microscopy for larger complexes

    • NMR spectroscopy for dynamic regions and solution behavior

  • Comparative Structural Analysis:

    • Superimpose L. monocytogenes nagB with structures from other organisms

    • Identify unique structural features of the serotype 4b enzyme

    • Map conserved catalytic residues and variable regulatory sites

    • Compare with E. coli nagB to understand similarities and differences in regulation

  • Molecular Dynamics Simulations:

    • Model conformational changes during catalysis

    • Simulate allosteric communication between binding sites

    • Predict effects of mutations on protein stability and function

    • Investigate protein flexibility and its relation to activity

  • Hydrogen-Deuterium Exchange Mass Spectrometry:

    • Map protein dynamics and conformational changes

    • Identify regions involved in allosteric regulation

    • Characterize binding interfaces with regulatory proteins

    • Detect subtle structural changes induced by different effectors

  • Site-Directed Spin Labeling and EPR Spectroscopy:

    • Measure distances between specific residues

    • Track conformational changes upon binding

    • Characterize protein dynamics in solution

    • Complement crystallographic data with solution-phase information

  • Integrative Structural Biology:

    • Combine multiple techniques (X-ray, NMR, SAXS, cryo-EM)

    • Build comprehensive models of protein complexes

    • Correlate structural features with functional data

    • Develop predictive models for rational enzyme engineering

This comprehensive structural biology approach can reveal the molecular basis for the complex regulation of nagB by multiple factors (GlcNAc-6P, HPr, NanE, and U-PII) and provide insights into how these interactions fine-tune enzyme activity in response to carbon and nitrogen availability . Understanding these mechanisms at the atomic level could inform the development of specific inhibitors targeting L. monocytogenes metabolism.

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.