Recombinant Bacillus subtilis Putative pyruvyl transferase epsO (epsO)

Shipped with Ice Packs
In Stock

Description

Molecular and Functional Characterization of EpsO

The epsO gene is part of the 15-gene epsA–epsO operon in B. subtilis, which is essential for biofilm formation . EpsO is annotated as a putative pyruvyl transferase, an enzyme responsible for transferring pyruvate groups to polysaccharides. This modification confers negative charges to EPS, enhancing biofilm stability and adhesion .

Key Functional Insights:

  • Biofilm Dependency: Strains lacking epsO exhibit defective biofilm formation, underscoring its role in EPS modification .

  • Enzymatic Activity: Pyruvyl transferases like EpsO catalyze the addition of pyruvate to carbohydrate polymers, altering their physicochemical properties .

  • Structural Prediction: EpsO is predicted to adopt a GT-B glycosyltransferase fold, common in enzymes involved in polysaccharide biosynthesis .

Recombinant Expression Systems for EpsO

Recombinant EpsO is produced using B. subtilis as a host due to its GRAS (Generally Recognized as Safe) status, efficient secretion machinery, and genetic tractability . Key strategies include:

Table 1: Expression Strategies for Recombinant Proteins in B. subtilis

ComponentDescriptionExample Application for EpsO
PromotersInducible (Pveg) or constitutive (P43) promoters drive gene expressionHigh-yield EpsO production
Signal PeptidesSec or Tat pathway peptides (e.g., SacB, XynA) enable extracellular secretionSecretion of soluble EpsO
Protease-Deficient StrainsStrains like WB800 (8 proteases deleted) reduce protein degradationEnhanced EpsO stability
Genome IntegrationChromosomal integration via amyE or lacA loci ensures genetic stabilityStable EpsO expression

Role in Biofilm Engineering

Recombinant EpsO has been utilized to study biofilm matrix composition. Pyruvate-modified EPS produced by EpsO increases biofilm resilience to environmental stressors, such as desiccation and antimicrobial agents .

Biotechnological Relevance

  • Material Science: Engineered biofilms incorporating EpsO-modified EPS serve as templates for durable living materials (e.g., silica composites) .

  • Industrial Enzymes: EpsO’s activity is leveraged in microbial factories for tailored polysaccharide production, such as hyaluronic acid derivatives .

Challenges and Optimizations

  • Secretion Efficiency: Screening signal peptides (e.g., LipA, CelA) improves EpsO translocation .

  • Proteolytic Degradation: Use of protease-deficient strains (e.g., WB800) increases yield .

  • Transcriptional Control: Riboswitch-regulated systems (e.g., theophylline-inducible P43) enable precise dose-dependent expression .

Future Directions

Further studies are needed to:

  • Resolve the crystal structure of EpsO to elucidate substrate-binding mechanisms.

  • Optimize fermentation conditions for industrial-scale production .

  • Explore synthetic biology tools (e.g., CRISPR-MAD7) for multiplexed eps locus engineering .

Product Specs

Form
Lyophilized powder. We preferentially ship the available format. If you have special format requirements, please note them when ordering.
Lead Time
Delivery time varies based on purchasing method and location. Consult local distributors for specifics. All proteins ship with blue ice packs by default. Request dry ice in advance for an extra fee.
Notes
Avoid repeated freeze-thaw cycles. Working aliquots can be stored at 4°C for up to one week.
Reconstitution
Briefly centrifuge the vial before opening. Reconstitute in sterile deionized water to 0.1-1.0 mg/mL. Add 5-50% glycerol (final concentration) and aliquot for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Liquid form: 6 months at -20°C/-80°C. Lyophilized form: 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. If you require a specific tag, please inform us for prioritized development.
Synonyms
epsO; yvfF; BSU34220Putative pyruvyl transferase EpsO; EC 2.-.-.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-322
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacillus subtilis (strain 168)
Target Names
epsO
Target Protein Sequence
MDSKHSMISL KQKLSGLLDV IPKQSEIIYA DYPLYGNVGD LFIMKGTEAF FKEHGIRVRK RWNPDNFPIG RKLDPNLIIV CQGGGNFGDL YPYYQGFREK IVQTYPNHKI VILPQSIYFQ NKDNLKRTAE IFSKHANLHI MTREKASYAT AQAYFTTNHI QLLPDMAHQL FPVIPTQQPS NQKLRFIRTD HEANQALQEH AEAESYDWRT VLSASDRRTI AFLQTLNVLN KKAGNPLPIA YIWEKYSDYI VQKAIRFFSR YESVETSRLH GHILSSLLQK ENTVIDNSYG KNANYFHTWM EGVPSTRLIQ HASKKENLPA HM
Uniprot No.

Target Background

Function
May contribute to exopolysaccharide (EPS) production in the extracellular matrix during biofilm formation. EPS facilitates the adhesion of cell chains into bundles.
Database Links
Protein Families
Polysaccharide pyruvyl transferase family

Q&A

What is the function of EpsO in Bacillus subtilis?

EpsO functions as a putative pyruvyl transferase involved in the production of the exopolysaccharide (EPS) component of B. subtilis extracellular matrix during biofilm formation. This enzyme belongs to the polysaccharide pyruvyl transferase family and is believed to catalyze the addition of pyruvyl groups to specific positions in the exopolysaccharide structure . The annotation of EpsO as a pyruvyltransferase is strongly supported by the presence of extremely conserved amino acids that are characteristic of this enzyme family . Within the biofilm context, the EPS produced by the epsA-O operon is responsible for the adhesion of chains of cells into bundles, which is essential for forming the three-dimensional architecture of B. subtilis biofilms . The pyruvylation of exopolysaccharides often confers specific physical properties to the polymer, including solubility, viscosity, and electrostatic interactions, which can significantly impact biofilm structure and stability.

The epsO gene is part of the epsA-O operon, which consists of 15 genes encoding proteins involved in exopolysaccharide biosynthesis and export. As one of the terminal enzymes in this pathway, EpsO likely acts on the polysaccharide after its initial assembly, adding chemical modifications that are crucial for the final functional properties of the EPS. Mutational studies of the eps operon have demonstrated that the absence of functional EPS production significantly impairs biofilm formation, highlighting the importance of this biosynthetic pathway and its components, including EpsO .

How does the epsA-O operon contribute to biofilm formation?

The epsA-O operon encodes 15 gene products that collectively synthesize the main extracellular polysaccharide in B. subtilis, which is essential for complex colony structure and pellicle formation . This operon is under the control of a sophisticated regulatory network whose primary function is to antagonize the SinR transcription repressor, thus derepressing the operons involved in matrix production during biofilm development . The EpsA-O exopolysaccharide produced by this operon forms a gel-like substance that spans the intercellular space, creating a complex three-dimensional biofilm network that provides structural support and protection to the bacterial community .

Experimental evidence clearly demonstrates the critical role of the epsA-O operon in biofilm development. When purified EpsA-O exopolysaccharide was exogenously added to Δeps mutant cells (which cannot produce their own EPS), the rheological behavior changed dramatically, with both storage and loss modulus increasing by an order of magnitude . More importantly, the qualitative behavior of the material transformed from a weak viscoelastic liquid to a viscoelastic solid, as indicated by a decrease in loss factor from 1.3 to 0.3 upon EpsA-O addition . This significant change in material properties highlights how the epsA-O operon products fundamentally alter the physical nature of bacterial aggregates, enabling them to form stable, structured biofilms rather than loose cell assemblages.

Within the operon, different genes serve various functions in EPS production. For instance, EpsA and EpsB are tyrosine kinases that regulate Eps production, while EpsC, EpsM, and EpsN are responsible for the biosynthesis of N,N′-diacetylbacillosamine (QuiNAc4NAc), a modified monosaccharide found in the EPS structure . EpsK functions as a membrane transporter similar to the Wzx protein of E. coli, and EpsL binds UDP-QuiNAc4NAc to the lipid acceptor undecaprenol phosphate . EpsO, as a putative pyruvyltransferase, contributes to the final modifications of the polysaccharide structure.

What is the structural characterization of EpsA-O exopolysaccharide?

The EpsA-O exopolysaccharide has been comprehensively characterized using chemical analysis, NMR spectroscopy, rheology, and molecular modeling techniques . These approaches have revealed a complex structure consisting of a trisaccharide backbone with the repeating unit [→3)-β-d-QuipNAc4NAc-(1→3)-β-d-GalpNAc-(1→3)-α-d-GlcpNAc-(1]n . This backbone is further elaborated with side chains composed of β-d-Galp(3,4-S-Pyr)-(1→6)-β-d-Galp(3,4-S-Pyr)-(1→6)-α-d-Galp-(1→ linked to C4 of GalNAc . The presence of pyruvyl ketal substituents, likely added by pyruvyltransferases such as EpsO, is a notable feature of this structure and contributes significantly to its physical properties.

The 1H NMR spectrum of native EpsA-O shows extremely broad lines, which persisted even after sonication, indicating the complex and potentially aggregating nature of this polysaccharide . Integration of the methyl region of the sonicated sample provided an approximate estimation of one deoxy-sugar (at 1.1 ppm), approximately 1.8 pyruvyl ketal substituents (at 1.6 ppm), and approximately four N-acetyl groups near 2 ppm . Chemical analysis detected only two N-acetylated sugars (GlcNAc and GalNAc), suggesting the presence of a 6-deoxy doubly N-acetylated residue, which was identified as QuiNAc4NAc .

The unique structure of the EpsA-O exopolysaccharide, particularly the presence of pyruvylated galactose residues in the side chains, contributes to its ability to form a gel-like matrix that provides structural support to the biofilm. The close agreement between the primary chemical structure and rheological behavior has allowed researchers to model the EpsA-O macromolecular and supramolecular solution structure, which demonstrates how this polysaccharide can span intercellular spaces to form a complex network .

How is the expression of epsO regulated during biofilm formation?

The expression of epsO, as part of the epsA-O operon, is regulated by a complex network of transcriptional controls that govern biofilm formation in B. subtilis. This regulatory network primarily functions to antagonize the SinR transcription repressor, thereby derepressing the operons involved in matrix production, including the epsA-O operon . SinR acts as a master regulator that normally represses biofilm formation during planktonic growth, but its activity is countered by anti-repressors such as SinI during conditions favorable for biofilm development.

The transition from planktonic to biofilm growth involves multiple environmental and cellular signals that feed into this regulatory network. While the search results do not provide specific details about epsO regulation in isolation, as part of the epsA-O operon, its expression would be coordinated with other genes in the operon to ensure the timely and appropriate production of exopolysaccharide components during biofilm formation. The expression of the entire eps operon is likely influenced by factors such as nutrient availability, cell density, and surface contact, which are known to trigger biofilm formation in B. subtilis.

Interestingly, research has shown that the role of SipW (a signal peptidase) in controlling matrix gene expression only occurs during biofilm formation on a solid surface, suggesting that different environmental contexts may employ distinct regulatory mechanisms for controlling eps operon expression . This environmental sensitivity highlights the sophisticated nature of the regulatory networks governing biofilm formation in B. subtilis, ensuring that resources are allocated to exopolysaccharide production only when conditions are appropriate for biofilm development.

What are the most effective methods for generating recombinant B. subtilis strains expressing modified epsO?

Generating recombinant B. subtilis strains with modified epsO requires careful consideration of expression systems, codon optimization, and transformation methods. One effective approach, based on methodologies described for other recombinant proteins in B. subtilis, involves first synthesizing the epsO gene using B. subtilis-preferred codons to optimize expression efficiency . Codon optimization is particularly important as it can significantly enhance protein expression by accommodating the codon usage bias of the host organism. The optimized gene sequence can then be inserted into a suitable expression vector such as plasmid pMK4, which has been successfully used for recombinant protein expression in B. subtilis .

For introducing specific modifications to the epsO gene, site-directed mutagenesis can be performed prior to insertion into the expression vector. This technique allows for precise alterations to the nucleotide sequence, enabling researchers to study the effects of specific amino acid changes on EpsO function. Following vector construction, transformation of B. subtilis can be achieved using standard competence protocols, and expression of the modified EpsO protein can be verified using Western blot analysis with appropriate antibodies .

To enhance the expression and secretion of recombinant EpsO, several engineering approaches can be employed. Co-expression of heterologous secretion machinery components, such as SecB from E. coli or DsbA from Staphylococcus, has been shown to potentially benefit protein secretion in B. subtilis . More extensive engineering approaches include modifications to the SecA protein, such as deletion in the C-terminal linker domain, which led to 2.2-fold enhanced secretion of human interferon-α2b in previous studies . Additionally, overexpression of the intramembrane protease RasP has demonstrated significant improvements in protein secretion, enhancing the secretion of certain enzymes by approximately 2.5- to 10-fold under industrial fermentation-mimicking conditions .

How can the pyruvyltransferase activity of EpsO be measured in vitro?

Measuring the pyruvyltransferase activity of EpsO in vitro requires establishing an assay system that can detect the transfer of pyruvyl groups from a donor molecule to an acceptor substrate. While specific methods for assaying EpsO activity are not detailed in the search results, several approaches can be adapted from general enzyme activity measurement techniques and knowledge of pyruvyltransferase biochemistry. The primary reaction catalyzed by pyruvyltransferases typically involves the transfer of a pyruvyl group from a donor such as phosphoenolpyruvate (PEP) to a specific position on a carbohydrate acceptor molecule.

One approach would be to develop a spectrophotometric assay that monitors the consumption of PEP or the formation of the pyruvylated product. For example, the decrease in absorbance at 240 nm can be used to track PEP consumption, while coupling the reaction to other enzymatic processes that produce detectable products could provide an indirect measure of activity. Alternatively, radiometric assays using 14C-labeled PEP could be employed to quantify the incorporation of radiolabeled pyruvyl groups into the acceptor substrate, providing a sensitive and direct measurement of EpsO activity.

Mass spectrometry-based methods offer another powerful approach for assessing pyruvyltransferase activity. High-resolution mass spectrometry can detect the mass shift corresponding to the addition of pyruvyl groups to the acceptor substrate, while tandem mass spectrometry (MS/MS) can provide structural information about the specific sites of pyruvylation. This is particularly valuable for characterizing the regiospecificity of EpsO, determining whether it adds pyruvyl groups to specific positions on the galactose residues in the EpsA-O side chains, as observed in the structural analysis of the exopolysaccharide .

For kinetic analysis of EpsO activity, enzyme assays would need to be conducted under varying substrate concentrations to determine parameters such as Km, Vmax, and kcat. This would provide insights into the catalytic efficiency of EpsO and its substrate preference. Additionally, assaying EpsO activity under different pH, temperature, and ionic strength conditions would help establish the optimal conditions for enzyme function, which could be valuable for both in vitro biochemical studies and biotechnological applications.

What are the structural implications of EpsO mutations on biofilm architecture?

Mutations in epsO could significantly impact the structure and properties of the B. subtilis exopolysaccharide, with consequent effects on biofilm architecture. Since EpsO is believed to catalyze the addition of pyruvyl groups to galactose residues in the side chains of the EpsA-O exopolysaccharide , mutations affecting its catalytic activity would likely result in reduced pyruvylation of the polysaccharide. The pyruvyl groups introduce negative charges and structural constraints to the polysaccharide, which influence its physical properties including solubility, viscosity, and interaction with other biofilm components.

Rheological studies have demonstrated that the EpsA-O exopolysaccharide plays a crucial role in determining the mechanical properties of B. subtilis biofilms. When purified EpsA-O was added to Δeps mutant cells, the material properties changed dramatically from a weak viscoelastic liquid to a viscoelastic solid, with both storage and loss modulus increasing by an order of magnitude . This suggests that mutations in epsO that reduce pyruvylation could lead to altered viscoelastic properties of the biofilm matrix, potentially decreasing its structural integrity and cohesiveness.

The complex three-dimensional architecture of B. subtilis biofilms depends on the ability of the exopolysaccharide to form a gel-like matrix that connects cells and provides structural support. Molecular modeling based on the chemical structure of EpsA-O has revealed how this polysaccharide can span intercellular spaces to create a complex network . Mutations in epsO that alter the chemical structure of the exopolysaccharide would likely disrupt this network formation, potentially leading to flatter, less structured biofilms with reduced resistance to mechanical stress and environmental challenges.

Beyond mechanical properties, changes in exopolysaccharide structure due to epsO mutations could affect biofilm functions such as adhesion to surfaces, resistance to antimicrobial agents, and protection from environmental stresses. The negative charges introduced by pyruvyl groups can influence interactions with metal ions and other charged molecules, potentially altering the biofilm's ability to sequester nutrients or toxic compounds. Additionally, the altered exopolysaccharide structure might affect interactions with other biofilm matrix components, such as the proteins TasA, TapA, and BslA, as well as extracellular DNA, which collectively contribute to biofilm structure and function .

How does EpsO interact with other proteins in the eps operon during exopolysaccharide synthesis?

The interaction of EpsO with other proteins in the eps operon is likely critical for coordinated exopolysaccharide synthesis, although the specific molecular interactions are not fully detailed in the search results. STRING database analysis reveals that EpsO has predicted functional partners, particularly EpsN, a putative aminotransferase also involved in EPS production, with a high confidence interaction score of 0.99 . This strong predicted interaction suggests that EpsO and EpsN may work closely together in the exopolysaccharide biosynthesis pathway, potentially forming a functional complex or acting sequentially in the modification of sugar residues.

The epsA-O operon encodes 15 proteins with diverse functions in exopolysaccharide biosynthesis, including regulatory proteins (EpsA, EpsB), enzymes involved in the synthesis of specialized sugars like QuiNAc4NAc (EpsC, EpsM, EpsN), membrane transporters (EpsK), and glycosyltransferases (EpsD) . As a putative pyruvyltransferase, EpsO likely functions toward the end of the biosynthetic pathway, adding pyruvyl groups to specific positions on the polysaccharide after its basic structure has been assembled. This would require coordination with proteins involved in earlier steps of the pathway to ensure that the appropriate substrate is available for pyruvylation.

The complexity of the EpsA-O exopolysaccharide structure, with its trisaccharide backbone and elaborated side chains containing pyruvylated galactose residues , implies a sophisticated biosynthetic machinery with multiple enzymes working in concert. The biosynthesis likely follows a sequential process where the backbone is synthesized first, followed by the addition of side chains and then chemical modifications such as pyruvylation. EpsO would therefore need to interact with or recognize the products of earlier biosynthetic steps, potentially through specific protein-protein interactions or by recognizing specific structural features of the partially synthesized polysaccharide.

Understanding the interactions between EpsO and other Eps proteins would be valuable for elucidating the complete pathway of exopolysaccharide biosynthesis in B. subtilis. Techniques such as co-immunoprecipitation, bacterial two-hybrid assays, or fluorescence resonance energy transfer (FRET) could be employed to identify direct protein-protein interactions. Additionally, structural studies of EpsO alone and in complex with potential interacting partners would provide insights into the molecular mechanisms underlying these interactions and how they contribute to coordinated exopolysaccharide synthesis.

What are the comparative differences between EpsO and other bacterial pyruvyltransferases?

Pyruvyltransferases are found in various bacterial species and play important roles in the modification of polysaccharides, contributing to the diverse structures and functions of bacterial capsules, exopolysaccharides, and cell wall components. While specific comparisons between EpsO and other bacterial pyruvyltransferases are not provided in the search results, the annotation of EpsO as a pyruvyltransferase is strongly supported by the presence of extremely conserved amino acids , indicating shared catalytic or structural features with other members of this enzyme family.

Bacterial pyruvyltransferases generally catalyze the transfer of a pyruvyl group from phosphoenolpyruvate to specific hydroxyl groups of carbohydrate residues, typically resulting in the formation of a ketal linkage. These enzymes often show specificity for both the sugar residue and the position of pyruvylation. For instance, the structural analysis of the EpsA-O exopolysaccharide revealed pyruvylation at positions 3 and 4 of galactose residues in the side chains , suggesting that EpsO has specificity for these positions. Comparative sequence analysis of EpsO with well-characterized pyruvyltransferases from other bacteria, such as WcaK from E. coli or CsaB from Bacillus anthracis, could reveal conserved catalytic residues and substrate-binding motifs.

The substrate specificity of pyruvyltransferases often reflects the unique polysaccharide structures produced by different bacterial species. For example, some pyruvyltransferases act on capsular polysaccharides, while others, like EpsO, modify exopolysaccharides involved in biofilm formation. The specific sugar residues targeted for pyruvylation can also vary, with some enzymes acting on mannose, galactose, or glucose residues. Understanding these specificities is important for predicting the functional roles of pyruvyltransferases in different bacterial species and potentially exploiting them for biotechnological applications.

Structural studies of pyruvyltransferases have revealed conserved domains and catalytic mechanisms, although the three-dimensional structure of EpsO itself has not been reported. Many pyruvyltransferases belong to the GT-B fold family of glycosyltransferases, featuring two Rossmann-like domains with the active site located in the cleft between them. Comparative modeling of EpsO based on the structures of related enzymes could provide insights into its catalytic mechanism and substrate binding mode, complementing biochemical and genetic studies to fully characterize this important enzyme in B. subtilis exopolysaccharide synthesis.

What protocols are most effective for purifying recombinant EpsO protein while maintaining enzymatic activity?

Purification of recombinant EpsO requires careful consideration of expression systems, purification tags, and buffer conditions to preserve enzymatic activity throughout the purification process. Based on approaches used for other recombinant proteins in B. subtilis, an effective protocol would begin with constructing an expression vector containing the epsO gene fused to a purification tag, such as a polyhistidine (His) tag or a Strep-tag II . These affinity tags facilitate efficient purification while typically having minimal impact on protein structure and function. The expression vector would be transformed into an appropriate B. subtilis strain, possibly one with enhanced protein secretion capabilities through overexpression of components like the intramembrane protease RasP, which has been shown to enhance protein secretion by 2.5- to 10-fold in industrial fermentation-mimicking conditions .

Following expression, cells would be harvested and lysed under mild conditions to preserve enzyme activity. For a putative membrane-associated enzyme like EpsO, careful selection of detergents for solubilization is crucial. Non-ionic detergents such as Triton X-100 or n-dodecyl-β-D-maltoside (DDM) at low concentrations often provide a good balance between effective solubilization and preservation of enzymatic activity. The initial purification step would typically involve affinity chromatography using the appropriate resin for the selected tag (e.g., Ni-NTA for His-tagged proteins). Following elution from the affinity column, the protein could be further purified by size exclusion chromatography to remove aggregates and obtain a homogeneous preparation.

Throughout the purification process, it's essential to monitor enzyme activity using an appropriate assay, such as those discussed in section 2.2. This allows for optimization of buffer conditions (pH, salt concentration, presence of stabilizers) to maintain activity. For pyruvyltransferases, considerations might include the addition of divalent cations like Mg2+ or Mn2+, which often serve as cofactors, and the inclusion of reducing agents like DTT or β-mercaptoethanol to maintain the redox state of any critical cysteine residues. Storage conditions should also be optimized, with options including flash freezing in liquid nitrogen with cryoprotectants, lyophilization, or storage at 4°C with appropriate stabilizers.

For structural and functional studies requiring higher purity, additional purification steps might be necessary. Ion exchange chromatography can be effective for removing contaminants with different charge properties, while hydrophobic interaction chromatography might separate proteins based on surface hydrophobicity. If the affinity tag might interfere with enzyme activity or structural studies, it can be removed using a specific protease (e.g., TEV protease for a TEV cleavage site) followed by an additional affinity chromatography step to separate the tag-free protein from the protease and uncleaved protein.

How can CRISPR-Cas9 be utilized to create precise mutations in epsO for functional studies?

CRISPR-Cas9 technology offers a powerful approach for introducing precise mutations in the epsO gene of B. subtilis, enabling detailed functional studies of this putative pyruvyltransferase. While specific CRISPR-Cas9 protocols for B. subtilis are not detailed in the search results, the general methodology can be adapted from established CRISPR-Cas9 genome editing approaches in bacteria. A comprehensive CRISPR-Cas9 strategy for epsO mutagenesis would begin with the design of guide RNAs (gRNAs) targeting specific regions of the epsO gene. These gRNAs would direct the Cas9 nuclease to create double-strand breaks at precise locations in the genome, which can then be repaired using homology-directed repair with a provided template containing the desired mutations.

For effective gRNA design, several considerations are important. First, target sequences should be unique in the B. subtilis genome to avoid off-target effects. Second, the target site should include a protospacer adjacent motif (PAM) sequence (typically NGG for Streptococcus pyogenes Cas9) immediately downstream of the target sequence. Third, gRNAs targeting regions near conserved amino acids that are likely involved in catalysis would be particularly valuable for functional studies of EpsO. Based on the annotation of EpsO as a putative pyruvyltransferase with extremely conserved amino acids , these conserved residues would be prime targets for mutagenesis.

In addition to the gRNA and Cas9 nuclease, a repair template containing the desired mutations must be designed. This template should include the mutation of interest flanked by homology arms (typically 500-1000 bp) matching the genomic sequence surrounding the cut site. For studying the function of specific amino acids in EpsO, point mutations changing catalytic residues to alanine or other amino acids could be introduced. Alternatively, domain swapping experiments could be designed by replacing segments of the epsO gene with corresponding regions from other pyruvyltransferases to investigate substrate specificity or catalytic efficiency.

Following transformation of B. subtilis with the CRISPR-Cas9 system components (gRNA, Cas9, and repair template), successful mutants can be screened using PCR amplification of the target region followed by sequencing. Colony PCR methods can be used for high-throughput screening of transformants. Once mutants are identified, their phenotypes should be characterized at multiple levels, including analysis of exopolysaccharide structure, biofilm formation capabilities, and pyruvyltransferase activity if purified enzyme assays are available. This comprehensive analysis would provide insights into the structure-function relationships of EpsO and its role in B. subtilis biofilm formation.

What spectroscopic methods are best suited for analyzing EpsO-catalyzed pyruvylation reactions?

Spectroscopic methods provide powerful tools for analyzing the pyruvyltransferase activity of EpsO and characterizing the products of its catalytic action. Based on the approaches used to characterize the EpsA-O exopolysaccharide structure, several spectroscopic techniques would be particularly valuable for studying EpsO-catalyzed reactions. Nuclear Magnetic Resonance (NMR) spectroscopy stands out as a particularly informative technique, as it was successfully employed to detect pyruvyl ketal substituents in the EpsA-O structure . 1H NMR can identify the characteristic methyl signal of pyruvyl groups at around 1.6 ppm, while 13C NMR can provide information about the ketal carbon and the positions of pyruvylation on the sugar residues.

For monitoring EpsO-catalyzed reactions in real-time or analyzing reaction kinetics, UV-visible spectroscopy could be employed if the reaction can be coupled to spectrophotometrically detectable changes. For example, if phosphoenolpyruvate (PEP) serves as the pyruvyl donor, its consumption could be monitored by the decrease in absorbance at 240 nm. Alternatively, coupling the pyruvyltransferase reaction to other enzymatic processes that generate or consume chromogenic or fluorogenic compounds would enable indirect monitoring of EpsO activity with high sensitivity.

Mass spectrometry offers another powerful approach for analyzing EpsO-catalyzed pyruvylation. Electrospray ionization mass spectrometry (ESI-MS) can detect the mass shift corresponding to the addition of pyruvyl groups to sugar residues or oligosaccharide substrates. For more detailed structural information, tandem mass spectrometry (MS/MS) can fragment the pyruvylated products to determine the specific positions of modification. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry might be particularly suitable for analyzing larger oligosaccharide substrates and products.

For studying the structural changes in the substrate upon pyruvylation, circular dichroism (CD) spectroscopy could be valuable, especially if the modification induces conformational changes in the polysaccharide. Additionally, Fourier-transform infrared (FTIR) spectroscopy could detect the characteristic vibrational modes of pyruvyl ketals and provide information about hydrogen bonding and other non-covalent interactions in the modified polysaccharide. Combining multiple spectroscopic techniques would provide complementary information about EpsO-catalyzed reactions, offering a comprehensive view of the enzyme's activity and the structural consequences of pyruvylation on the exopolysaccharide.

How can the contribution of EpsO to biofilm mechanical properties be quantitatively assessed?

Quantitatively assessing the contribution of EpsO to biofilm mechanical properties requires a combination of genetic approaches, material characterization techniques, and advanced imaging methods. Rheology has emerged as a particularly valuable technique for studying biofilm mechanical properties, as demonstrated by its successful application in characterizing the effects of EpsA-O exopolysaccharide on B. subtilis biofilms . To specifically assess the contribution of EpsO, comparative rheological analysis could be performed on biofilms formed by wild-type B. subtilis, epsO deletion mutants, and complemented strains expressing either wild-type or catalytically inactive EpsO.

The search results indicate that addition of purified EpsA-O exopolysaccharide to Δeps mutant cells dramatically changed their rheological behavior, with both storage modulus (G') and loss modulus (G") increasing by an order of magnitude, and the material transitioning from a weak viscoelastic liquid to a viscoelastic solid . Similar experiments could be conducted with exopolysaccharide purified from epsO mutants to determine how the absence of pyruvylation affects these mechanical properties. Oscillatory rheometry with frequency sweeps would provide information about the viscoelastic behavior across different timescales, while creep tests could assess the long-term deformation response under constant stress.

Beyond bulk rheology, microscale techniques can provide spatial information about biofilm mechanical properties. Atomic force microscopy (AFM) in force spectroscopy mode can measure the elastic modulus and adhesion forces at specific points within the biofilm, potentially revealing spatial heterogeneity in mechanical properties that might be influenced by EpsO activity. Microindentation using calibrated microbeads or specialized probes can also provide localized measurements of biofilm stiffness and viscoelasticity. These techniques could be particularly valuable for comparing the mechanical properties of surface layers versus deeper regions of biofilms formed by wild-type and epsO mutant strains.

The mechanical resistance of biofilms to fluid shear stress is another important property that could be influenced by EpsO activity. Flow chamber experiments with controlled shear rates could assess the ability of biofilms to withstand mechanical perturbation, with quantitative measures including the critical shear stress required for biofilm detachment or the rate of biomass loss under defined shear conditions. Additionally, confocal microscopy combined with image analysis techniques can provide quantitative measures of biofilm structural parameters such as thickness, surface roughness, and porosity, which may correlate with mechanical properties and be influenced by EpsO-mediated pyruvylation of the exopolysaccharide .

What are the recommended approaches for studying EpsO localization within B. subtilis cells?

Studying the subcellular localization of EpsO within B. subtilis cells requires specialized techniques to visualize and track this protein in living or fixed bacterial cells. One of the most widely used approaches involves creating translational fusions of EpsO with fluorescent proteins such as Green Fluorescent Protein (GFP) or its variants. These fusion constructs can be expressed from their native genetic locus to maintain physiological expression levels or from inducible promoters for higher expression when needed. Time-lapse fluorescence microscopy of B. subtilis expressing EpsO-GFP fusions would allow for dynamic tracking of protein localization during biofilm development, potentially revealing changes in distribution or abundance under different conditions or growth phases.

For higher-resolution imaging beyond the diffraction limit of conventional light microscopy, super-resolution techniques such as Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED) microscopy, or Single-Molecule Localization Microscopy (SMLM) could be employed. These approaches can resolve structures down to tens of nanometers, potentially allowing visualization of EpsO clustering or association with specific subcellular structures. Another powerful approach is Fluorescence Resonance Energy Transfer (FRET), which could be used to detect interactions between EpsO and other proteins involved in exopolysaccharide synthesis, such as its predicted functional partner EpsN .

Immunofluorescence microscopy using specific antibodies against EpsO represents another valuable approach, particularly if fluorescent protein fusions affect protein function or localization. This technique involves fixing and permeabilizing cells, then incubating them with primary antibodies against EpsO followed by fluorescently labeled secondary antibodies. While this approach does not allow live-cell imaging, it can provide high specificity and does not require genetic modification of the epsO gene. For even higher resolution, immunogold labeling combined with electron microscopy could visualize EpsO at the nanometer scale, potentially revealing its association with membrane structures or exopolysaccharide export machinery.

Biochemical approaches like subcellular fractionation followed by Western blotting can complement microscopy techniques by quantitatively assessing the distribution of EpsO across different cellular compartments (cytoplasm, membrane, cell wall, and extracellular fractions). Since EpsO is predicted to be involved in exopolysaccharide synthesis, it might localize to the cytoplasmic membrane or be associated with membrane complexes involved in polysaccharide export. Density gradient centrifugation could further separate different membrane domains, potentially revealing associations with specific lipid environments or multiprotein complexes involved in exopolysaccharide synthesis and export.

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.