Recombinant Listeria monocytogenes serotype 4b Glucosamine--fructose-6-phosphate aminotransferase [isomerizing] (glmS), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult 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 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 serves 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 have a 12-month shelf life 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 you require a specific tag, please inform us for preferential development.
Synonyms
glmS; LMOf2365_0762Glutamine--fructose-6-phosphate aminotransferase [isomerizing]; EC 2.6.1.16; D-fructose-6-phosphate amidotransferase; GFAT; Glucosamine-6-phosphate synthase; Hexosephosphate aminotransferase; L-glutamine--D-fructose-6-phosphate amidotransferase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
glmS
Uniprot No.

Target Background

Function
This enzyme catalyzes the initial step in hexosamine metabolism, converting fructose-6-phosphate to glucosamine-6-phosphate using glutamine as the nitrogen source.
Database Links
Subcellular Location
Cytoplasm.

Q&A

What is the function of the glmS enzyme in Listeria monocytogenes?

The glmS enzyme in Listeria monocytogenes encodes Glucosamine--fructose-6-phosphate aminotransferase [isomerizing], which catalyzes the first step in hexosamine biosynthesis. This essential enzyme converts fructose-6-phosphate to glucosamine-6-phosphate (GlcN6P), a critical precursor for bacterial cell wall biosynthesis. The GlmS enzyme is directly involved in the synthesis pathway of UDP-N-acetylglucosamine (UDP-GlcNAc), which is a fundamental building block for peptidoglycan and teichoic acids in the bacterial cell wall .

This enzymatic function is particularly important in Listeria monocytogenes, a Gram-positive bacterium that causes listeriosis, a severe foodborne illness with high mortality rates. The proper functioning of GlmS is essential for cell wall integrity, which impacts bacterial survival, especially under stress conditions such as exposure to lysozyme, an enzyme that can degrade bacterial cell walls .

How is glmS expression regulated in Listeria monocytogenes?

The glmS gene in Listeria monocytogenes is regulated by a unique RNA-based mechanism involving a ribozyme located in the 5' untranslated region (UTR) of the glmS mRNA. This regulatory element, known as the glmS ribozyme, functions as both a ribozyme (catalyzing RNA self-cleavage) and a riboswitch (binding specific metabolites to alter gene expression) .

The glmS ribozyme specifically binds glucosamine-6-phosphate (GlcN6P), the product of the GlmS enzyme reaction. When GlcN6P levels are high, this metabolite binds to the ribozyme, activating its self-cleavage activity. This cleavage destabilizes the mRNA, leading to its degradation and consequently reducing GlmS protein production. This creates a negative feedback loop that maintains appropriate levels of GlcN6P in the cell .

Research has shown that the L. monocytogenes glmS ribozyme has unique properties compared to ribozymes from other bacterial species, including stricter discrimination between phosphorylated and non-phosphorylated metabolites and the ability to maintain activity at low temperatures .

What experimental approaches are used to study glmS ribozyme activity?

Researchers employ several methodological approaches to characterize glmS ribozyme activity:

  • In vitro transcription and purification: RNA corresponding to the glmS ribozyme region is synthesized using in vitro transcription with T7 RNA polymerase and purified using denaturing polyacrylamide gel electrophoresis (PAGE) .

  • Metabolite-dependent self-cleavage assays: Purified ribozyme RNA is incubated with various concentrations of potential co-factors (e.g., GlcN6P, GlcN) under controlled conditions. The self-cleavage reaction is then analyzed to determine:

    • EC50 values (effective concentration for 50% cleavage)

    • Reaction kinetics

    • Temperature dependence

    • Divalent cation requirements

  • Structural analysis: Secondary structure prediction tools and comparative sequence analysis are used to determine the 2D structures of the ribozyme cores .

For the L. monocytogenes glmS ribozyme, these approaches have revealed an EC50 value of approximately 0.38 μM for GlcN6P, indicating high sensitivity compared to glmS ribozymes from other bacteria such as B. subtilis and S. aureus .

How does the L. monocytogenes glmS ribozyme differ from other bacterial glmS ribozymes in terms of co-factor specificity and environmental adaptations?

The L. monocytogenes glmS ribozyme demonstrates unique characteristics that distinguish it from other bacterial homologs. Experimental characterization reveals several key differences:

  • Co-factor specificity: The L. monocytogenes glmS ribozyme exhibits unprecedented stringent discrimination between phosphorylated and non-phosphorylated metabolites. It shows an EC50 value of 0.38 μM for GlcN6P compared to 1920 μM for GlcN, representing a 5000-fold difference in sensitivity. This discriminatory power is significantly higher than observed in other bacterial species .

  • Temperature adaptability: Unlike glmS ribozymes from other bacteria, the L. monocytogenes variant maintains efficient cleavage activity at temperatures as low as 6°C. This unique cold-tolerance property likely reflects an adaptation to L. monocytogenes' ability to grow at refrigeration temperatures, a trait that contributes to its persistence in cold food storage environments .

  • Sensitivity to GlcN6P: The L. monocytogenes glmS ribozyme demonstrates EC50 values in the nanomolar range, making it approximately 7 times more sensitive to GlcN6P than the S. aureus ribozyme and over 5000-fold more sensitive than the B. subtilis variant .

  • Structural differences: Analysis of nucleotide sequences reveals differences in the P1 loop regions, with the L. monocytogenes ribozyme having a 39-nucleotide loop compared to 12 nt for B. subtilis, 27 nt for C. difficile, and 62 nt for S. aureus. These structural variations may contribute to the observed differences in cleavage rates and metabolite sensitivity .

This exceptional selectivity for phosphorylated metabolites likely involves the conserved Mg2+ ion complex, which plays a crucial role in high-affinity binding through interaction with the phosphate group of GlcN6P .

What is the relationship between GlmS and GlmR in L. monocytogenes, and how does this impact bacterial virulence?

The relationship between GlmS and GlmR in L. monocytogenes represents a crucial aspect of cell wall biosynthesis regulation with direct implications for virulence:

  • Functional relationship: While previous studies in Bacillus subtilis suggested a direct protein-protein interaction between GlmR and GlmS that enhances GlmS activity, research in L. monocytogenes reveals a different mechanism. Despite both proteins being capable of homodimerization (as demonstrated through bacterial two-hybrid assays), no direct interaction between L. monocytogenes GlmR and GlmS has been detected .

  • Metabolic role: Instead of directly modulating GlmS activity, L. monocytogenes GlmR functions as an accessory uridyltransferase that catalyzes the synthesis of UDP-GlcNAc from UTP and N-acetylglucosamine-1-phosphate. This enzymatic activity contributes to the cellular pool of UDP-GlcNAc, a critical cell wall precursor .

  • Virulence implications: GlmR deficiency in L. monocytogenes leads to:

    • Decreased resistance to cell-wall stress (particularly lysozyme)

    • Reduced cytosolic survival in host cells

    • Impaired ability to avoid inflammasome activation

    • Significantly attenuated virulence in vivo

These phenotypes are linked to reduced UDP-GlcNAc levels in ΔglmR mutants, as demonstrated by untargeted metabolomics analysis .

Importantly, suppressor mutations that block non-essential wall teichoic acid decoration pathways (which normally consume UDP-GlcNAc) can partially restore lysozyme resistance and virulence in ΔglmR mutants. This indicates that the primary function of GlmR in L. monocytogenes is to ensure adequate UDP-GlcNAc synthesis for essential cell wall components .

What experimental approaches are most effective for studying recombinant GlmS from L. monocytogenes serotype 4b?

For researchers working with recombinant L. monocytogenes serotype 4b GlmS, several methodological approaches have proven effective:

  • Recombinant protein expression and purification:

    • Cloning the glmS gene into expression vectors with appropriate affinity tags (His6, GST)

    • Expression in E. coli systems (BL21(DE3) or similar strains)

    • Induction optimization (IPTG concentration, temperature, duration)

    • Purification using affinity chromatography followed by size exclusion chromatography

    • Verification of protein purity using SDS-PAGE and Western blotting

  • Enzymatic activity assays:

    • Spectrophotometric assays measuring the conversion of fructose-6-phosphate to glucosamine-6-phosphate

    • Coupled enzyme assays tracking the consumption of glutamine or production of glutamate

    • Kinetic analysis to determine Km, Vmax, and catalytic efficiency

    • Inhibition studies to identify potential antimicrobial compounds

  • Structural biology approaches:

    • X-ray crystallography to determine three-dimensional structure

    • Site-directed mutagenesis to identify catalytic residues

    • Molecular dynamics simulations to understand conformational changes

  • Interaction studies:

    • Bacterial two-hybrid assays to investigate potential protein-protein interactions

    • Pull-down assays to identify binding partners in cellular extracts

    • Surface plasmon resonance to quantify binding affinities

When studying the relationship between GlmS and the glmS ribozyme, researchers should consider:

  • RNA footprinting to map ribozyme-metabolite interactions

  • In vitro transcription-translation assays to monitor regulation

  • Reporter gene fusions to track gene expression in vivo

How do mutations in the glmS gene affect L. monocytogenes virulence and antimicrobial resistance?

Mutations in the glmS gene can have profound effects on L. monocytogenes virulence and antimicrobial resistance due to the critical role of GlmS in cell wall biosynthesis:

  • Impact on cell wall integrity:

    • Reduced GlmS activity leads to decreased glucosamine-6-phosphate production

    • This results in weakened peptidoglycan structure and altered teichoic acid composition

    • The compromised cell wall increases susceptibility to osmotic stress, cell wall-targeting antimicrobials, and host defense mechanisms

  • Effects on virulence:

    • GlmS is essential for survival in the host cytosol

    • Mutations affecting GlmS function reduce bacterial persistence in macrophages

    • Defects in cell wall synthesis trigger stronger inflammasome activation, increasing host immune recognition

    • These factors collectively attenuate virulence in animal infection models

  • Antimicrobial resistance implications:

    • Mutations reducing GlmS activity increase susceptibility to lysozyme and other cell wall-targeting antimicrobials

    • Conversely, adaptive mutations that enhance glmS expression can potentially increase resistance to certain antibiotics

    • The glmS ribozyme represents a potential target for novel antimicrobial strategies

Interestingly, compensatory mechanisms exist that can partially restore virulence in strains with defects in the GlmS pathway. For example, mutations that block non-essential pathways consuming UDP-GlcNAc (such as wall teichoic acid decoration) can redirect the limited pool of this precursor toward essential cell wall components, partially restoring lysozyme resistance and virulence .

What are the optimal conditions for expressing and purifying recombinant L. monocytogenes GlmS?

Successful expression and purification of recombinant L. monocytogenes GlmS requires careful optimization of multiple parameters:

  • Expression system selection:

    • E. coli BL21(DE3) is commonly used for GlmS expression

    • Consider specialized strains for potentially toxic proteins (C43(DE3), Rosetta for rare codons)

    • Evaluate both N- and C-terminal tag placements, as tag position can affect folding and activity

  • Expression conditions optimization:

    • Temperature: Lower temperatures (16-25°C) often improve solubility

    • IPTG concentration: Typically 0.1-0.5 mM, with lower concentrations promoting soluble expression

    • Expression duration: 4-16 hours depending on temperature and construct

  • Buffer optimization for purification:

    • Base buffer: 50 mM Tris-HCl or phosphate buffer (pH 7.5-8.0)

    • Salt concentration: 150-300 mM NaCl to maintain solubility

    • Glycerol (5-10%) to enhance stability

    • Reducing agents (1-5 mM DTT or β-mercaptoethanol) to prevent disulfide formation

    • Protease inhibitors during initial lysis steps

  • Purification strategy:

    • Initial capture: Immobilized metal affinity chromatography (IMAC) for His-tagged constructs

    • Intermediate purification: Ion exchange chromatography (typically anion exchange)

    • Polishing: Size exclusion chromatography

    • Tag removal consideration: Include a TEV or thrombin protease site if tag-free protein is needed

  • Stability considerations:

    • Addition of 5-10% glycerol to storage buffer

    • Flash-freezing in liquid nitrogen in small aliquots

    • Storage at -80°C for long-term preservation

A typical purification protocol yields approximately 5-10 mg of purified GlmS per liter of bacterial culture, with >95% purity as assessed by SDS-PAGE.

How can researchers effectively analyze the interaction between recombinant GlmS and the glmS ribozyme?

Investigating the complex relationship between GlmS enzyme activity and glmS ribozyme regulation requires specialized approaches:

  • In vitro transcription of glmS ribozyme:

    • Design DNA templates containing the T7 promoter followed by the complete ribozyme sequence

    • Include precise 5' and 3' boundaries based on secondary structure predictions

    • Perform transcription in buffer containing 40 mM Tris-HCl (pH 7.9), 10 mM NaCl, 6 mM MgCl2, 2 mM spermidine, 10 mM DTT, and 2 mM of each NTP

    • Purify transcripts using denaturing PAGE

  • Self-cleavage activity assays:

    • Pre-fold ribozyme RNA (1-2 pmol) in reaction buffer

    • Initiate cleavage by adding various concentrations of GlcN6P (0.01 μM to 10 mM)

    • Quench reactions at various time points using formamide loading buffer

    • Analyze cleavage products by denaturing PAGE and phosphorimaging

    • Calculate EC50 values and reaction rates

  • Feedback loop analysis:

    • Set up coupled in vitro transcription-translation systems containing:

      • Purified recombinant GlmS

      • Template DNA with glmS gene including its native ribozyme

      • Necessary cofactors and substrates

    • Monitor protein synthesis using Western blotting or reporter systems

    • Analyze ribozyme cleavage products by Northern blotting

  • Cellular studies:

    • Develop reporter gene constructs fused to the glmS ribozyme

    • Introduce these constructs into L. monocytogenes alongside manipulations of GlmS levels

    • Monitor reporter expression under various conditions

    • Correlate with measurements of cellular GlcN6P levels

Table 1: Comparison of EC50 values for glmS ribozymes from different bacterial species

Bacterial SpeciesEC50 for GlcN6P (μM)EC50 for GlcN (μM)Discrimination FactorTemperature Range for Activity
L. monocytogenes0.38192050006-37°C
C. difficile0.46Not determinedNot determined25-37°C
S. aureus~2.7~600~22025-37°C
B. subtilis>1000>10000~1025-37°C

This comparative data highlights the exceptional sensitivity and discriminatory power of the L. monocytogenes glmS ribozyme, particularly its unparalleled ability to function at low temperatures .

What genomic approaches can be used to study the conservation and evolution of glmS in different L. monocytogenes strains?

Genomic approaches provide valuable insights into the conservation, variation, and evolution of the glmS gene across L. monocytogenes strains and serotypes:

  • Whole-genome sequencing and comparative genomics:

    • Next-generation sequencing of diverse L. monocytogenes isolates

    • Core-genome multi-locus sequence typing (cgMLST) to determine phylogenetic relationships

    • Analysis of single nucleotide polymorphisms (SNPs) in the glmS coding region and regulatory elements

    • Assessment of selection pressures using dN/dS ratio calculations

  • Ribozyme sequence and structure analysis:

    • Multiple sequence alignment of glmS ribozyme regions across strains

    • Secondary structure prediction and comparison

    • Identification of conserved nucleotides essential for ribozyme function

    • Detection of compensatory mutations maintaining RNA structure

  • Transcriptomic approaches:

    • RNA-seq analysis to compare glmS expression levels across strains

    • Differential expression analysis under various growth conditions

    • Identification of transcription start sites and processing events

    • Detection of small RNAs potentially regulating glmS expression

  • Population genomics:

    • Analysis of glmS sequence variation in epidemiologically diverse isolates

    • Correlation of specific variants with clinical vs. environmental sources

    • Assessment of temporal persistence of specific glmS genotypes

    • Identification of mutations associated with enhanced virulence or stress resistance

Research has shown that while the glmS coding sequence is highly conserved across L. monocytogenes strains, subtle variations exist in the ribozyme region that may influence the efficiency of metabolite sensing and self-cleavage. Additionally, the genetic context surrounding the glmS gene can vary between different clonal complexes (CCs), potentially affecting its regulation and expression levels .

How can understanding the glmS system contribute to the development of novel antimicrobial strategies against L. monocytogenes?

The unique characteristics of the L. monocytogenes glmS system offer several promising avenues for antimicrobial development:

  • Ribozyme-targeted approaches:

    • Design of GlcN6P analogs that trigger ribozyme self-cleavage but cannot be metabolized

    • Development of small molecules that stabilize the cleaved state of the ribozyme

    • Creation of antisense oligonucleotides targeting critical ribozyme structures

    • Screening for compounds that interfere with ribozyme-metabolite interactions

  • GlmS enzyme inhibition strategies:

    • Structure-based design of competitive inhibitors blocking the active site

    • Allosteric inhibitors disrupting enzyme dimerization or conformational changes

    • Covalent inhibitors targeting catalytic residues

    • Exploration of natural products with GlmS-inhibitory activity

  • GlmR-targeted approaches:

    • Inhibition of GlmR uridyltransferase activity to limit UDP-GlcNAc synthesis

    • Disruption of potential regulatory interactions between GlmR and other cellular components

    • Development of combination therapies targeting both GlmS and GlmR pathways

  • Metabolic intervention strategies:

    • Manipulation of intracellular GlcN6P levels to disrupt feedback regulation

    • Development of GlcN6P analogs that compete for ribozyme binding without triggering cleavage

    • Targeting downstream pathways dependent on UDP-GlcNAc

The exceptional discriminatory power of the L. monocytogenes glmS ribozyme between phosphorylated and non-phosphorylated metabolites offers opportunities for highly specific therapeutic interventions with potentially reduced off-target effects on human cells or beneficial microbiota.

What are the current challenges in studying the structure-function relationship of L. monocytogenes GlmS?

Researchers face several significant challenges when investigating the structure-function relationships of L. monocytogenes GlmS:

  • Structural complexity:

    • GlmS is a large, multi-domain enzyme requiring sophisticated structural biology approaches

    • The presence of flexible regions complicates crystallization efforts

    • The homodimeric nature adds complexity to structural analysis

    • Obtaining structures with bound substrates or inhibitors remains challenging

  • Functional analysis limitations:

    • The essential nature of glmS makes genetic manipulation difficult

    • Conditional mutants are often needed to study function in vivo

    • Separating enzyme function from ribozyme regulatory effects requires careful experimental design

    • Differences between in vitro and in vivo activity can complicate interpretation

  • Technical challenges:

    • Producing sufficient quantities of active, properly folded recombinant protein

    • Developing sensitive and specific assays for GlmS activity

    • Accurately measuring intracellular metabolite levels (particularly GlcN6P)

    • Monitoring ribozyme activity in cellular contexts

  • Translational barriers:

    • Balancing antimicrobial potency with specificity

    • Achieving sufficient cellular penetration of potential inhibitors

    • Addressing potential for resistance development

    • Demonstrating efficacy in relevant infection models

Despite these challenges, advances in cryo-electron microscopy, metabolomics, and genetic manipulation techniques are gradually enabling more detailed analyses of the GlmS structure-function relationship in L. monocytogenes.

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.