Recombinant Enterococcus faecalis Serine hydroxymethyltransferase (glyA)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Enterococcus faecalis Serine Hydroxymethyltransferase (glyA)

Serine hydroxymethyltransferase (SHMT), encoded by the glyA gene, is an enzyme crucial for cellular metabolism, catalyzing the reversible conversion of serine and tetrahydrofolate to glycine and 5,10-methylene-tetrahydrofolate . In Enterococcus faecalis, a bacterium known for its role as both a commensal and an opportunistic pathogen, SHMT plays a vital role in various metabolic processes . The recombinant form of E. faecalis SHMT (rEf-SHMT) allows for detailed studies of its structure, function, and potential applications .

Role in Enterococcus faecalis Metabolism

SHMT (glyA) is integral to the bacterium's survival and adaptation within its environment . Its functions include:

  • Glycine biosynthesis SHMT provides glycine, a precursor for proteins, nucleotides, and other essential biomolecules.

  • Folate metabolism By producing 5,10-methylene-tetrahydrofolate, SHMT contributes to the folate cycle, vital for nucleotide synthesis and DNA methylation.

  • Nutrient acquisition E. faecalis can degrade N-linked glycans of human RNase B to acquire nutrients .

  • Biofilm formation Glycosyltransferases (GTF) play roles in biofilm-associated antibiotic resistance, cell envelope integrity, and conjugative transfer .

Recombinant Production and Purification

Producing rEf-SHMT involves cloning the glyA gene from E. faecalis into an expression vector, transforming it into a suitable host strain (e.g., E. coli), and inducing protein expression . The expressed protein is then purified using affinity chromatography, often employing a His-tag fused to the rEf-SHMT.

Table 1: Steps for Recombinant Production and Purification of rEf-SHMT

StepDescription
CloningAmplify the glyA gene from E. faecalis genomic DNA and insert it into an expression vector.
TransformationIntroduce the expression vector into a suitable host strain, such as E. coli.
ExpressionInduce protein expression by adding IPTG or another suitable inducer to the culture medium.
Cell LysisHarvest the cells and lyse them to release the expressed protein.
PurificationUse affinity chromatography (e.g., Ni-NTA) to purify the rEf-SHMT based on its His-tag.
DialysisRemove any residual salts or impurities from the purified protein.
Quality ControlConfirm the purity and activity of the rEf-SHMT using SDS-PAGE and enzyme activity assays.

Research Applications and Findings

The availability of rEf-SHMT has facilitated numerous research investigations:

  • Enzyme kinetics and mechanism Detailed kinetic studies have elucidated the catalytic mechanism of E. faecalis SHMT, providing insights into substrate binding, transition state stabilization, and product release.

  • Inhibitor design Knowledge of the enzyme's structure and mechanism has aided the design of potential inhibitors targeting SHMT, which could have implications for developing novel antimicrobial agents.

  • Structural studies X-ray crystallography and other biophysical techniques have been employed to determine the three-dimensional structure of rEf-SHMT, offering a visual representation of the active site and potential allosteric regulatory sites.

  • Glycan degradation E. faecalis O-Glycosidase removes O-glycans from glycoproteins and is broadly active on Core-1, Core-2, Core-3, and Gal-Core-2 structures .

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 purchasing method and location. Please contact your local distributor for precise delivery estimates. Note: Standard shipping includes 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 settle 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 default glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on 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 for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. The tag type will be determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
glyA; EF_2550Serine hydroxymethyltransferase; SHMT; Serine methylase; EC 2.1.2.1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-412
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Enterococcus faecalis (strain ATCC 700802 / V583)
Target Names
glyA
Target Protein Sequence
MDYKTYDPDL WNAIAREEER QENNLELIAS ENVVSKAVMA AQGSILTNKY AEGYPGKRYY GGCEFIDIVE NLAIDRAKEL FGAKFANVQA HSGSQANTAA YLSLVEPGDT ILGMDLSAGG HLTHGSPVNF SGKTYNFVSY GVDPSTEVID YDVVRILARE HRPKLIVAGA SAYSRTIDFK RFREIADEVD AKLMVDMAHI AGLVASGLHP NPVPYADIVT STTHKTLRGP RGGLILTNSE ELAKKVNSSI FPGIQGGPLE HVIAGKAAAF KEALDPSFAE YSQQVIANAQ AMTKVFNQAP EARLISGATD NHLLLIEVTG FGLNGKEAEA ILDSVNITVN KNSIPFEQLS PFKTSGIRIG TPAITSRGFK EEDAVEVAKL IVQVLKDPEN TAVHDEVKAA VAALTKKYPL YN
Uniprot No.

Target Background

Function

Serine hydroxymethyltransferase (glyA) catalyzes the reversible interconversion of serine and glycine, utilizing tetrahydrofolate (THF) as a one-carbon carrier. This reaction is the primary source of one-carbon units crucial for the biosynthesis of purines, thymidylate, methionine, and other essential biomolecules. It also exhibits THF-independent aldolase activity towards beta-hydroxyamino acids, producing glycine and aldehydes via a retro-aldol mechanism.

Database Links

KEGG: efa:EF2550

STRING: 226185.EF2550

Protein Families
SHMT family
Subcellular Location
Cytoplasm.

Q&A

What is the function of serine hydroxymethyltransferase (SHMT) in Enterococcus faecalis?

SHMT, encoded by the glyA gene, is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that catalyzes the reversible interconversion of serine and tetrahydrofolate (THF) to glycine and 5,10-methylene tetrahydrofolate (MTHF). This reaction is crucial as MTHF is a major source of cellular one-carbon units necessary for biosynthesis of purines and thymidylate . In E. faecalis, this enzyme plays a vital role in one-carbon metabolism that is essential for bacterial viability and pathogenicity. Unlike in eukaryotes, SHMT in bacterial systems like E. faecalis typically exists as a single homodimeric enzyme .

Why is the glyA gene considered essential in bacterial systems?

The glyA gene is considered essential in many bacterial systems because its product (SHMT) is pivotal for:

  • One-carbon metabolism necessary for nucleotide biosynthesis

  • Production of glycine, an essential amino acid for protein synthesis

  • Generation of MTHF, which is critical for DNA synthesis and cellular replication

In studies of minimal bacterial genomes, such as those performed with JCVI-syn3A, glyA consistently appears as an essential gene that cannot be removed without severely compromising bacterial viability. Transposon mutagenesis and FBA (Flux Balance Analysis) models confirm that the removal of glyA results in non-viability or severe growth impairment . Studies with H. pylori demonstrated that a ΔglyA strain could be obtained but exhibited markedly slowed growth (21 hours doubling time versus 4 hours for wild-type) and lost virulence factors like CagA .

How is recombinant E. faecalis SHMT typically produced for research purposes?

Recombinant E. faecalis SHMT can be produced through several expression systems:

E. coli expression system:

  • The glyA gene is amplified from E. faecalis genomic DNA using PCR with primers containing appropriate restriction sites

  • The gene is cloned into expression vectors like pET21b or pQE30 that provide His-tags for purification

  • Expression is induced in E. coli with IPTG, and the protein is purified using affinity chromatography

Example protocol from literature:
For cytoplasmic expression in complementation assays, glyA can be amplified with primers containing appropriate restriction sites (e.g., NdeI and NotI) and cloned into pET21b. For periplasmic production, the gene can be cloned into vectors like pASK-IBA2c to generate an N-terminal OmpA-leader peptide fused, C-terminal Strep-tagged protein .

Purification conditions typically involve buffers containing:

  • 2% N-lauroylsarcosine (or 0.1% in washing buffer)

  • 2 mM 1,4-dithiothreitol (DTT)

  • 50 μM PLP as a cofactor

What expression systems are available for recombinant protein production in E. faecalis?

Several inducible expression systems have been developed for E. faecalis:

  • Agmatine-inducible system (pAGEnt vector):

    • Utilizes the agmatine deiminase (AGDI) pathway components

    • Contains the aguR inducer gene and the aguB promoter

    • Induction with 60 mM agmatine results in high expression levels

    • Shows close correlation between inducer concentration and expression level

  • Nisin-inducible system (NICE):

    • Originally developed for Lactococcus lactis but adapted for Enterococcus

    • Requires regulatory genes nisRK either in trans or on the same plasmid

    • Well-established system for controlled gene expression

  • Other systems:

    • Rhamnose-inducible system

    • Pheromone cCF10-controlled expression system

The choice depends on experimental needs, with the agmatine-inducible system offering the advantage of being a single-plasmid expression system requiring no additional regulatory genes in trans .

How can knockout mutants of glyA be generated in E. faecalis, and what compensatory mechanisms enable viability?

Creating glyA knockout mutants in E. faecalis requires sophisticated approaches due to the gene's essentiality:

1. RecT recombinase-mediated gene editing with CRISPR-Cas9:
The most efficient approach involves:

  • Expressing E. faecalis RecT recombinase to improve recombineering efficiency

  • Co-transformation with single-stranded DNA template and pCas9 plasmid targeting the wild-type locus

  • Selection using appropriate antibiotics (chloramphenicol for Cas9 expression)

  • Screening for successful recombination events

This method has shown significant improvements over traditional approaches, with transformation efficiency reaching up to 10⁵ CFU/μg DNA in some Enterococcus strains .

  • Alternative pathways for glycine biosynthesis (e.g., threonine aldolase activity)

  • Uptake of extracellular glycine through amino acid transporters

  • Metabolic rewiring to bypass one-carbon metabolism requirements

  • Suppressor mutations that activate alternative pathways

In H. pylori, a ΔglyA strain could be obtained but with severe growth defects (doubling time increased from 4 to 21 hours). Similar compensatory mechanisms likely operate in E. faecalis .

What experimental designs are most effective for studying the role of glyA in E. faecalis virulence?

Robust experimental designs for studying glyA's role in virulence should incorporate:

1. True experimental designs with proper controls:

  • Pretest-posttest control group design (Design #4 in Campbell & Stanley's classification)

  • Solomon four-group design to account for testing effects (Design #5)

2. In vitro virulence assays:

  • Adhesion to epithelial cell lines (e.g., Caco-2)

  • Biofilm formation capacity measurements

  • Growth in various nutrient-limited media

  • Resistance to oxidative stress and antimicrobial peptides

3. Parallel and crossover encouragement designs:
These designs allow for the manipulation of mediator variables when direct manipulation is difficult:

  • Randomized encouragement to take certain values of mediators after treatment

  • Enables informative inferences about causal mechanisms by focusing on subsets of the population

4. Animal infection models with defined endpoints:

  • Mouse peritonitis model (survival rates at 100 hours post-infection)

  • UTI models (kidney colonization measurements)

  • Endocarditis models (vegetation formation)

5. Molecular characterization:

  • Transcriptomics to identify networks affected by glyA disruption

  • Metabolomics to trace one-carbon unit flux

  • Proteomics to examine changes in virulence factor expression

A comprehensive approach would include both complementation studies (restoring wild-type phenotype with functional glyA) and dose-dependent expression systems to correlate glyA levels with virulence traits.

How does PLP binding affinity affect the enzymatic activity of E. faecalis SHMT, and what structural features contribute to this?

PLP binding is critical for SHMT activity, and research has revealed interesting characteristics in bacterial SHMTs:

Binding characteristics:

  • H. pylori SHMT shows unexpectedly weak binding affinity for PLP compared to other bacterial SHMTs

  • The three-dimensional structure of H. pylori SHMT apoprotein determined at 2.8Å resolution suggests structural features contributing to this low affinity

  • Similar studies in E. faecalis SHMT have identified two variable loops crucial for inhibitor binding

  • Serine binding to SHMT enhances the affinity of inhibitors by stabilizing the loop structure

Structural features affecting PLP binding:

  • The active site is typically located at the interface between two monomers

  • Key residues involved in PLP binding are highly conserved across species

  • The conformation of mobile loops can significantly affect cofactor binding and catalysis

  • X-ray crystallography studies (PDB: 7V3D) have revealed specific structural details of E. faecium SHMT in complex with inhibitors

Methodological approaches to study PLP binding:

  • Spectroscopic evidence can indicate formation of enzyme-PLP-glycine-folate complexes

  • Biochemical assays can measure the effects of varying PLP concentrations on enzyme activity

  • Site-directed mutagenesis can identify critical residues for PLP binding

  • Structural biology approaches (X-ray crystallography, cryo-EM) provide atomic-level details

These insights have implications for drug design, as stabilization of inactive configurations using small molecules has potential for specifically inhibiting bacterial SHMT without affecting human homologs .

How can recombinant E. faecalis SHMT be used as a tool to study one-carbon metabolism in bacterial systems?

Recombinant E. faecalis SHMT serves as a valuable tool for studying one-carbon metabolism through several approaches:

1. Metabolic flux analysis:

  • Isotope labeling experiments using ¹³C-serine or ¹³C-glycine

  • Tracking labeled carbon atoms through metabolic networks using mass spectrometry

  • Quantifying flux through the folate cycle and connected pathways

2. Enzyme kinetics and substrate specificity:

  • Determining kinetic parameters (Km, kcat, kcat/Km) for different substrates

  • Comparing parameters between bacterial and human SHMT to identify metabolic differences

  • Exploring THF-independent aldolytic cleavage, decarboxylation, and transamination reactions catalyzed by SHMT

3. Protein-protein interaction studies:

  • Identifying interaction partners using pull-down assays or yeast two-hybrid screening

  • Characterizing multi-enzyme complexes involved in one-carbon metabolism

  • Mapping metabolic channeling mechanisms between enzymes

4. Inhibitor screening and characterization:

  • High-throughput screening for SHMT inhibitors

  • Structure-activity relationship studies

  • Evaluation of synergistic effects with other metabolic inhibitors

Research has demonstrated that SHMT inhibitors like (+)-SHIN-1 can synergistically enhance the antibacterial activities of nucleoside analogues, highlighting the interconnected nature of one-carbon metabolism with nucleotide synthesis pathways .

What are the most advanced techniques for measuring and characterizing glycosylation of recombinant proteins in E. faecalis?

E. faecalis contains glycosylation machinery that can modify recombinant proteins. Advanced techniques to study these modifications include:

1. Mass spectrometry-based approaches:

  • UHPLC coupled with mass spectrometry to identify released glycoforms

  • Glycopeptide analysis using electron transfer dissociation (ETD) fragmentation

  • Intact protein mass spectrometry to determine glycoform distributions

2. Lectin blot analysis:

  • Using various lectins with different specificities to detect specific glycan structures

  • Comparative analysis before and after enzymatic deglycosylation

3. Enzymatic characterization:
Research has identified glycosyltransferases involved in protein glycosylation in E. faecalis:

  • The gtfAB locus (gtfA and gtfB; EF2891 and EF2892 in E. faecalis V583) shares the same glycosyl transferase domain (PFAM PF00534)

  • In-frame deletion of this locus prevents glycosylation of target proteins

  • Effect of glycosylation on protein function can be assessed by measuring bacterial chain length using flow cytometry and microscopy

4. Recombinant expression of E. faecalis glycosidases:

  • The EndoE enzyme from E. faecalis has been identified as an endo-β-N-acetylglucosaminidase

  • It hydrolyzes N-linked glycans from human glycoproteins like IgG and lactoferrin

  • These enzymes can be used as tools to characterize glycan structures

5. Functional characterization:

  • Assessing how glycosylation affects protein folding, stability and function

  • Determining the role of glycans in immune evasion and host-pathogen interactions

  • Investigating the impact on biofilm formation and virulence

The study of glycosylation in E. faecalis has revealed roles in septum cleavage during growth, bacterial chain length regulation, and potentially in host-pathogen interactions .

What are the key considerations for designing complementation assays to verify glyA function?

Designing robust complementation assays for glyA requires careful consideration of several factors:

1. Host strain selection:

  • Use E. coli strains with glyA deletion (e.g., E. coli ΔglyA)

  • Ensure the strain has glycine auxotrophy phenotype

  • Consider using strains with additional features like λ phage redγβα operon for recombination

2. Vector design:

  • Include appropriate promoters (IPTG-inducible promoters like T7 or tac)

  • Add affinity tags for protein detection and purification (His-tag, Strep-tag)

  • Incorporate proper signal sequences for intended cellular localization

3. Growth conditions:

  • Minimal media compositions that restrict glycine availability

  • Media supplemented with serine to assess SHMT's function in serine-to-glycine conversion

  • Various induction conditions (inducer concentration, temperature, duration)

4. Controls:

  • Positive control: wild-type strain with functional glyA

  • Negative control: deletion strain transformed with empty vector

  • Additional controls: strains expressing known functional or non-functional SHMT variants

5. Phenotypic assessment:

  • Growth curve measurements in minimal media with and without glycine

  • Colony formation on solid minimal medium

  • Measurement of glycine production by HPLC or other analytical methods

Example protocol from literature:
The complementation ability of H. pylori SHMT was assessed using an E. coli ΔglyA strain transformed with plasmid pQE60 carrying the IPTG-inducible ORF HP0183. Tests were performed on solid minimal M9 medium in the presence or absence of glycine and serine. The wild-type E. coli MG1665 and the deletion strain transformed with pQE60 without insert served as controls. Growth on minimal medium in the presence of IPTG demonstrated successful complementation .

How can researchers address the technical challenges of working with glycosyltransferases in Enterococcus faecalis?

Working with glycosyltransferases in E. faecalis presents several technical challenges that can be addressed through specialized approaches:

1. Gene identification and characterization:

  • Use bioinformatic approaches to identify putative glycosyltransferases based on conserved domains (e.g., PFAM PF00534)

  • Verify function through complementation studies and enzymatic assays

  • Research has identified the gtfAB locus (EF2891 and EF2892 in E. faecalis V583) as key glycosyltransferases involved in protein glycosylation

2. Expression systems:

  • Utilize the agmatine-inducible expression system (pAGEnt vector) for controlled expression

  • Consider periplasmic expression using signal sequences for proper protein folding

  • Use fusion partners to enhance solubility

3. Enzyme activity assays:

  • Develop in vitro assays using synthetic acceptor substrates

  • Utilize radiolabeled or fluorescently labeled sugar donors

  • Employ mass spectrometry to detect and characterize glycosylation products

4. Protein modification detection:

  • Use TEV protease cleavage followed by analysis to detect glycosylated peptides

  • Employ lectin blotting with different lectins to detect specific glycan structures

  • Apply mass spectrometry techniques for detailed glycan characterization

5. Functional verification:

  • Construct deletion mutants (ΔgtfAB) using allele exchange

  • Compare phenotypes between wild-type and mutant strains

  • Assess effects on cellular processes like septum cleavage using flow cytometry and microscopy

Research has shown that deletion of glycosyltransferases in E. faecalis affects bacterial chain length, with ΔgtfAB mutants forming longer chains (10-20 cells) compared to wild-type strains (2-4 cells), indicating their role in cell division processes .

What statistical approaches are most appropriate for analyzing gene essentiality data for glyA in different experimental contexts?

Statistical analysis of gene essentiality data requires specialized approaches depending on the experimental context:

1. Transposon mutagenesis data analysis:

  • Use statistical models that account for insertion biases

  • Apply Hidden Markov Models to identify essential genomic regions

  • Compare insertion frequencies between genes to establish essentiality thresholds

  • In genome-scale transposon mutagenesis studies, genes with no or significantly fewer insertions than expected are classified as essential

2. Flux Balance Analysis (FBA) in silico predictions:

  • Statistical comparison between model-derived and experimental gene essentiality

  • Calculation of true positive rate, false positive rate, and Matthews correlation coefficient

  • Identification of discrepancies that suggest knowledge gaps in metabolic networks

  • Studies with JCVI-syn3A showed good agreement between transposon- and model-derived gene essentiality, with every in silico essential gene being at least quasi-essential in vivo

3. Growth phenotype analysis:

  • ANOVA or mixed-effects models for growth curve data

  • Survival analysis for time-to-growth measurements

  • Nonparametric tests when assumptions of normality are violated

  • In H. pylori, ΔglyA strains showed significantly impaired growth (doubling time 21 hours vs. 4 hours for wild-type)

4. Gene expression data in complementation studies:

  • Multiple testing correction using False Discovery Rate (FDR) control

  • Methods for estimating the true number of differentially expressed genes

  • Consider the mixture of P-values between differentially and non-differentially expressed genes

5. Experimental design considerations:

  • Power analysis to determine appropriate sample sizes

  • Randomization, replication, and blocking to control for confounding factors

  • Use of quasi-experimental designs when full randomization is not possible

For microarray or RNA-seq experiments, it's crucial to understand that even with FDR control at 20%, only a small fraction of truly differentially expressed genes may be detected when replication is low. Methods like that of Langaas et al. can be used to estimate the number of differentially expressed genes and assess the extent of type 2 errors .

How can structural information about E. faecalis SHMT be leveraged for antimicrobial drug development?

Structural information about E. faecalis SHMT provides valuable insights for antimicrobial drug development:

1. Structure-based drug design approaches:

  • Crystal structures of bacterial SHMTs, such as the complex structure of serine hydroxymethyltransferase from E. faecium (PDB: 7V3D), reveal detailed binding sites for inhibitor design

  • Identification of two variable loops crucial for inhibitor binding

  • Understanding that serine binding enhances inhibitor affinity by stabilizing loop structure

2. Targeting bacterial-specific features:

  • Comparative analysis between bacterial and human SHMT structures

  • Identification of unique pockets or conformations in bacterial enzymes

  • Focusing on differences in oligomeric state (bacterial SHMTs are typically homodimeric while human cytosolic SHMT is tetrameric)

3. Developing synergistic approaches:

  • Research has shown that SHMT inhibitors like (+)-SHIN-1 can bacteriostatically inhibit the growth of E. faecium at remarkably low concentrations (EC₅₀ of 10⁻¹¹M)

  • These inhibitors synergistically enhance the antibacterial activities of nucleoside analogues

  • This synergism provides a strategy to overcome resistance mechanisms

4. Exploiting PLP binding mechanisms:

  • Studies of H. pylori SHMT revealed unexpectedly weak binding affinity for PLP

  • Structural basis for this low affinity can guide inhibitor design

  • Stabilization of inactive configurations using small molecules has potential for specific inhibition

5. Virtual screening and optimization:

  • Molecular docking studies using bacterial SHMT structures

  • Fragment-based approaches to identify initial binding scaffolds

  • Molecular dynamics simulations to account for protein flexibility

The development of SHMT inhibitors has broader implications beyond antibacterial therapies, with potential applications for treating bacterial, viral, and parasitic infections as well as cancer .

What is the potential role of E. faecalis SHMT in biofilm formation and how might this be exploited therapeutically?

E. faecalis SHMT plays several potential roles in biofilm formation that could be exploited therapeutically:

1. Metabolic contributions to biofilm matrix:

  • One-carbon metabolism provides building blocks for nucleotide synthesis

  • These nucleotides contribute to extracellular DNA, a major component of biofilm matrix

  • Inhibition of SHMT could disrupt this supply chain, weakening biofilm structure

2. Interaction with host glycoproteins:

  • E. faecalis produces EndoE, an endo-β-N-acetylglucosaminidase that hydrolyzes N-linked glycans from human glycoproteins

  • Human lactoferrin (hLF) inhibits biofilm formation of E. faecalis in vitro

  • EndoE-hydrolyzed hLF inhibits biofilm formation to a lesser extent than intact hLF

  • This suggests EndoE prevents inhibition of biofilm by modifying host glycoproteins

3. Nutrient acquisition in biofilm environment:

  • Culture experiments showed that EndoE enables E. faecalis to use glycans derived from lactoferrin as a carbon source

  • Similar mechanisms might involve SHMT in alternative carbon metabolism pathways

  • This could be crucial for bacterial survival in nutrient-limited biofilm environments

4. Therapeutic approaches:

  • SHMT inhibitors could disrupt biofilm formation and increase susceptibility to antibiotics

  • Combining SHMT inhibitors with glycosidase inhibitors might have synergistic effects

  • Targeting both metabolic and structural components of biofilms simultaneously

5. Connection to virulence:

  • Biofilm formation is linked to virulence and antibiotic resistance

  • Understanding SHMT's role could provide targets for anti-virulence therapies

  • This approach might reduce selective pressure for resistance development

Research on E. faecalis glycobiology has demonstrated that glycans play crucial roles in the interplay between bacteria and the human host, with implications for both commensalism and opportunistic pathogenicity . Targeting these interactions could provide novel therapeutic approaches.

How does the genomic context of glyA in different E. faecalis strains correlate with pathogenicity and antibiotic resistance profiles?

The genomic context of glyA varies across E. faecalis strains and correlates with pathogenicity and antibiotic resistance in several ways:

1. Genetic conservation and variation:

2. Genomic neighborhood analysis:

  • In some bacterial operons, glyA is part of multigene operons

  • In H. pylori 26695, the glyA open reading frame (HP0183) is part of an operon with non-essential ORFs (HP0184 and HP0185) located downstream

  • Similar operon structures in E. faecalis may influence regulation and expression patterns

3. Correlation with antibiotic resistance:

  • The epidemiological shift in hospitals from E. faecalis to E. faecium has been linked to antibiotic resistance patterns

  • Currently, 90% and 80% of E. faecium from healthcare-associated infections are resistant to ampicillin and vancomycin, respectively

  • While E. faecalis remains largely susceptible to these antibiotics

  • These differences in susceptibility patterns may be influenced by metabolic adaptations involving one-carbon metabolism

4. Correlation with pathogenicity:

  • In some bacterial species, glyA deletion mutants show attenuated virulence

  • For example, E. ictaluri glyA deletion mutants were significantly attenuated in virulence

  • In V. cholerae, SHMT (GlyA1) was identified as a MetR-regulated virulence factor required for intestinal colonization

5. Strain classification and epidemiology:

  • Human-associated E. faecium are phylogenetically separated into two distinct clades

  • Clade A1 represents pathogenic strains, while Clade B represents commensal strains

  • Genetic tools for studying glyA function need to be adapted for different clades

  • For example, vancomycin-resistant E. faecium are commonly naturally resistant to erythromycin, requiring alternative selection markers for genetic manipulation

Understanding these correlations can inform surveillance efforts, guide antibiotic stewardship practices, and help identify high-risk strains based on genetic signatures.

What are the optimal conditions for purifying enzymatically active recombinant E. faecalis SHMT?

Purifying enzymatically active recombinant E. faecalis SHMT requires careful attention to several factors:

1. Expression conditions:

  • Host strain: E. coli BL21(DE3) or similar expression strains

  • Induction: Low IPTG concentration (0.1-0.5 mM) at lower temperatures (16-25°C)

  • Addition of cofactors: 50-100 μM PLP in the growth medium

  • Supplementation with 200 μM folinic acid can improve folding and activity

2. Buffer composition for purification:

  • Inclusion of PLP (50 μM) in all purification buffers to maintain cofactor binding

  • Addition of reducing agents (2 mM DTT or 5 mM β-mercaptoethanol)

  • Mild detergents (0.1-2% N-lauroylsarcosine) may help maintain solubility

  • pH range of 7.5-8.0 for optimal stability

3. Purification strategy:
For His-tagged proteins:

  • Use Ni-NTA affinity chromatography with imidazole gradients

  • Follow with size exclusion chromatography to separate oligomeric states

  • Consider ion exchange chromatography as a polishing step

For Strep-tagged proteins:

  • Use the protocol for cleared lysates as recommended by manufacturers

  • Buffers should contain 2% N-lauroylsarcosine (or 0.1% in washing/elution buffers)

  • Include 2 mM DTT and 50 μM PLP in all buffers

4. Quality control:

  • SDS-PAGE to confirm purity

  • Spectroscopic analysis to verify PLP binding (characteristic absorption at 425 nm)

  • Specific activity measurements using standard SHMT assays

  • Thermal shift assays to confirm proper folding

5. Storage conditions:

  • Store with 10-20% glycerol at -80°C for long-term storage

  • Avoid repeated freeze-thaw cycles

  • Consider flash-freezing in small aliquots

Example protocol from literature:
E. coli JM83 cells transformed with pASK-IBA2c-glyA were grown in no salt LB with 30 μg/ml chloramphenicol, 250 mM sucrose, and 50 mM L-serine at 30°C. After induction at OD600 of 1.2 with 200 ng/ml anhydrotetracyline, 50 μM PLP and 200 μM folinic acid were added followed by 4h incubation at 25°C. Purification used manufacturer-recommended protocols with modifications including 2% N-lauroylsarcosine in buffers (0.1% in washing/elution buffers), 2 mM DTT, and 50 μM PLP .

What are the most reliable assays for measuring SHMT enzymatic activity in E. faecalis extracts?

Several reliable assays are available for measuring SHMT activity in E. faecalis extracts:

1. Spectrophotometric assays:

a) Coupled enzyme assay with NADH oxidation:

  • SHMT reaction coupled to 5,10-methylene-THF dehydrogenase

  • Measures NADH oxidation at 340 nm

  • Advantage: Continuous monitoring of reaction

  • Limitation: Interference from other NADH-consuming activities

b) Direct UV-based assay:

  • Measures absorbance changes at 240 nm due to methenyl-THF formation

  • Advantage: Simplicity

  • Limitation: Lower sensitivity and potential interference

2. Radiochemical assays:

a) [¹⁴C]-Serine to [¹⁴C]-glycine conversion:

  • Incubation of enzyme with [¹⁴C]-serine and THF

  • Separation of products by paper chromatography or HPLC

  • Quantification by scintillation counting

  • Advantage: High sensitivity and specificity

  • Limitation: Requires radioactive facilities

3. HPLC-based assays:

a) Separation and quantification of reaction products:

  • Derivatization of amino acids using o-phthalaldehyde or similar reagents

  • Reverse-phase HPLC separation

  • Fluorescence detection

  • Advantage: High sensitivity and simultaneous analysis of multiple metabolites

  • Limitation: Time-consuming

4. Specialized activity assays:

a) DAAO coupled enzymatic assay for racemase activity:

  • Detects D-alanine produced by SHMT racemase activity

  • D-Ala is deaminated into pyruvate by D-amino acid oxidase (DAAO)

  • Pyruvate is quantified using 2,4-dinitrophenylhydrazine (DNPH)

  • Advantage: Specific for racemase activity

  • Limitation: Indirect measurement

b) Functional complementation assays:

  • Uses E. coli ΔglyA strains

  • Measures growth restoration on minimal media

  • Advantage: Demonstrates physiological relevance

  • Limitation: Qualitative rather than quantitative

When selecting an assay, researchers should consider sensitivity requirements, available equipment, and potential interfering activities in E. faecalis extracts. For complex samples, controls for non-specific activities are essential.

What strategies can be employed to improve solubility and stability of recombinant E. faecalis SHMT?

Improving solubility and stability of recombinant E. faecalis SHMT requires multiple strategies:

1. Expression optimization:

a) Temperature modulation:

  • Lower induction temperatures (16-20°C)

  • Slow induction using auto-induction media

  • Heat shock (42°C for 20 min) before lowering to expression temperature

b) Codon optimization:

  • Adjust codon usage for expression host

  • Remove rare codons that may cause translational pausing

c) Co-expression with chaperones:

  • GroEL/GroES, DnaK/DnaJ/GrpE systems

  • Trigger factor to aid co-translational folding

2. Protein engineering approaches:

a) Fusion partners:

  • Solubility enhancers: MBP, SUMO, Trx, GST, NusA

  • Inclusion of cleavable linkers for tag removal

  • C-terminal rather than N-terminal tags to allow proper folding

b) Surface engineering:

  • Identification and mutation of surface hydrophobic patches

  • Introduction of surface salt bridges to enhance stability

  • Glycine to alanine substitutions in flexible regions

3. Buffer optimization:

a) Stabilizing additives:

  • Pyridoxal 5'-phosphate (PLP) at 50-100 μM in all buffers

  • Osmolytes: glycerol (10-20%), sucrose (5-10%), trehalose

  • Reducing agents: DTT (1-5 mM), β-mercaptoethanol, TCEP

  • Mild detergents: N-lauroylsarcosine (0.1-2%), Triton X-100

b) pH and salt optimization:

  • Systematic screening of pH ranges (typically 7.0-8.5)

  • NaCl concentration screening (100-500 mM)

  • Addition of divalent cations (Mg²⁺, Ca²⁺)

4. Purification strategies:

a) Rapid processing:

  • Minimize time between cell lysis and purification

  • Cold room operations

  • Addition of protease inhibitors

b) Specialized techniques:

  • On-column refolding for inclusion bodies

  • Size exclusion chromatography to remove aggregates

  • Affinity chromatography in the presence of stabilizing ligands

5. Storage conditions:

a) Cryoprotectants:

  • Glycerol (20-50%)

  • Sucrose or trehalose (10-20%)

b) Formulation:

  • Flash-freezing in small aliquots

  • Lyophilization with appropriate excipients

  • Storage at -80°C rather than -20°C

Example from literature: For purification of Chlamydia pneumoniae SHMT, researchers used a buffer containing 2% N-lauroylsarcosine (reduced to 0.1% in washing/elution buffers), 2 mM DTT, and 50 μM PLP, which significantly improved protein solubility and stability .

How can researchers accurately quantify and compare the expression levels of glyA in different E. faecalis strains or growth conditions?

Accurate quantification of glyA expression requires multiple complementary techniques:

1. Quantitative real-time PCR (qRT-PCR):

a) Protocol considerations:

  • RNA extraction using specialized kits for Gram-positive bacteria

  • DNase treatment to remove genomic DNA contamination

  • Reverse transcription with random primers or gene-specific primers

  • qPCR with validated primer pairs targeting glyA

b) Data analysis:

  • Normalization with multiple reference genes (e.g., 16S rRNA, rpoB, gyrA)

  • Use of the 2⁻ΔΔCt method for relative quantification

  • Absolute quantification using standard curves if needed

2. RNA-Seq analysis:

a) Experimental design:

  • Include biological triplicates for each condition

  • Deep sequencing (>10 million reads per sample)

  • Strand-specific libraries to distinguish antisense transcription

b) Bioinformatic analysis:

  • Quality control and adapter trimming

  • Mapping to reference genome using STAR or HISAT2

  • Quantification using featureCounts or HTSeq

  • Differential expression analysis with DESeq2 or edgeR

  • Apply appropriate statistical methods with FDR correction

3. Protein-level quantification:

a) Western blotting:

  • Generation of specific antibodies against E. faecalis SHMT

  • Careful optimization of extraction conditions

  • Inclusion of loading controls (e.g., GroEL, DnaK)

  • Densitometric analysis for semi-quantitative comparison

b) Mass spectrometry-based proteomics:

  • Label-free quantification using spectral counting or intensity-based methods

  • Targeted proteomics using selected/multiple reaction monitoring (SRM/MRM)

  • Absolute quantification using AQUA peptides or QconCAT standards

4. Reporter gene assays:

a) Promoter-reporter fusions:

  • Cloning glyA promoter upstream of fluorescent proteins (GFP, mCherry)

  • Measurement of fluorescence using plate readers or flow cytometry

  • Enables single-cell analysis of expression heterogeneity

b) Using existing expression systems:

  • The agmatine-inducible system with GFP reporter has shown close correlation between inducer concentration and fluorescence

  • Under induction with 60 mM agmatine, fluorescence reached 40 arbitrary units compared to 0 in uninduced cells

5. Considerations for experimental design:

a) Growth phase standardization:

  • Sampling at defined optical densities rather than time points

  • Use of chemostats for steady-state expression analysis

b) Environmental controls:

  • Precise temperature control (±0.5°C)

  • Media lot consistency

  • pH monitoring and control

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.