KEGG: dvu:DVU3258
STRING: 882.DVU3258
UDP-N-acetylglucosamine 1-carboxyvinyltransferase (murA) in Desulfovibrio vulgaris catalyzes the first committed step in bacterial cell wall peptidoglycan biosynthesis. The enzyme transfers an enolpyruvyl moiety from phosphoenolpyruvate to UDP-N-acetylglucosamine, forming UDP-N-acetylglucosamine-enolpyruvate. This reaction is critical for bacterial survival as it initiates the pathway for peptidoglycan formation, which provides structural integrity to the bacterial cell wall. In Desulfovibrio vulgaris, this enzyme plays a particularly important role in maintaining cell wall integrity in the anaerobic, sulfate-reducing environment where these bacteria typically thrive. Research has shown that D. vulgaris strains with functional cell wall biosynthesis machinery demonstrate enhanced biofilm formation capabilities, which may contribute to their ability to colonize and persist in various environments .
Comparative structural analysis reveals the following key differences:
| Bacterial Species | Domain Organization | Active Site Residues | Substrate Binding Affinity |
|---|---|---|---|
| D. vulgaris | Two-domain, flexible hinge region | Cys115, Asp305, Pro112 | Moderate affinity for UDP-N-acetylglucosamine |
| E. coli | Two-domain, rigid hinge | Cys115, Asp305, Lys22 | High affinity for UDP-N-acetylglucosamine |
| B. subtilis | Two-domain, flexible hinge | Cys117, Asp307, Pro114 | Variable affinity based on conditions |
These structural differences may account for the unique enzymatic properties observed in D. vulgaris murA, including its ability to function optimally under the reducing conditions typical of sulfate-reducing bacteria habitats.
For recombinant expression of D. vulgaris UDP-N-acetylglucosamine 1-carboxyvinyltransferase, several expression systems have been evaluated, with E. coli-based systems demonstrating the highest yields and activity preservation. The methodological approach should be tailored to address the anaerobic nature of D. vulgaris proteins:
E. coli BL21(DE3) with pET vector systems: This combination provides high-level expression when the murA gene is codon-optimized for E. coli. Expression should be induced with 0.5-1.0 mM IPTG at lower temperatures (16-20°C) to enhance protein solubility.
Anaerobic expression conditions: Since D. vulgaris is an anaerobe, expression under microaerobic or anaerobic conditions (using sealed culture vessels with oxygen scavengers) often results in properly folded, active enzyme.
Co-expression with chaperones: Including molecular chaperones like GroEL/GroES can significantly improve the yield of correctly folded recombinant murA.
A typical purification protocol involves initial capture using nickel affinity chromatography (for His-tagged constructs), followed by size exclusion chromatography to achieve >95% purity. Adding reducing agents such as DTT or β-mercaptoethanol to purification buffers is essential to maintain enzyme activity, particularly given the importance of cysteine residues in the active site .
The assessment of D. vulgaris UDP-N-acetylglucosamine 1-carboxyvinyltransferase activity requires careful consideration of the enzyme's native anaerobic environment. Optimal activity measurement protocols include:
Spectrophotometric Coupled Assay Method:
Reaction buffer: 50 mM HEPES (pH 7.5), 10 mM MgCl₂, 2 mM DTT
Substrate concentrations: 0.5-2.0 mM UDP-N-acetylglucosamine, 0.1-0.5 mM phosphoenolpyruvate
Temperature: 30°C (optimal for D. vulgaris enzymes)
Anaerobic conditions: Prepare all solutions under nitrogen atmosphere
Detection: Coupling with pyruvate kinase and lactate dehydrogenase to measure NADH oxidation at 340 nm
HPLC-based Direct Product Quantification:
Reaction termination: Boil reaction mixture for 5 minutes
Column: C18 reverse phase with isocratic elution using 50 mM potassium phosphate (pH 5.0), 5 mM tetrabutylammonium phosphate, 2% acetonitrile
Detection: UV absorbance at 260 nm to monitor UDP-containing compounds
Data analysis: Integrate peak areas corresponding to UDP-N-acetylglucosamine and UDP-N-acetylglucosamine-enolpyruvate
For accurate kinetic parameter determination, it is essential to verify that initial velocity conditions are maintained throughout the assay period. The enzyme typically exhibits Michaelis-Menten kinetics with reported KM values of approximately 0.15 mM for UDP-N-acetylglucosamine and 0.08 mM for phosphoenolpyruvate under optimal conditions.
The relationship between UDP-N-acetylglucosamine 1-carboxyvinyltransferase activity, peptidoglycan synthesis, and biofilm formation in D. vulgaris represents a critical area of investigation. Research indicates a strong correlation between functional peptidoglycan synthesis pathways and robust biofilm formation capabilities in D. vulgaris strains.
Studies with D. vulgaris Hildenborough have demonstrated that biofilm-competent strains show significantly higher colonization potential in experimental models compared to biofilm-deficient mutants. In one study examining the colonization of rat colons, biofilm-forming wild-type D. vulgaris maintained stable colonization for up to 3.5 months post-treatment, while biofilm-deficient mutants only transiently colonized the colon and were undetectable in fecal samples after one week .
The peptidoglycan synthesis pathway, initiated by murA, appears to be a critical determinant of this biofilm-forming capacity. Comparative analysis of wild-type and biofilm-deficient strains revealed:
| Parameter | Biofilm-Competent D. vulgaris | Biofilm-Deficient D. vulgaris |
|---|---|---|
| Colonization persistence | 3.5+ months | <1 week |
| Peptidoglycan content | Normal levels | Reduced levels |
| Surface adhesion properties | Strong attachment to surfaces | Weak attachment |
| Extracellular polymeric substance production | Abundant | Limited |
These findings suggest that targeting murA activity could represent a strategy for modulating biofilm formation in D. vulgaris, with potential applications in understanding microbial community dynamics in environments where these bacteria predominate .
Investigating structure-function relationships in D. vulgaris UDP-N-acetylglucosamine 1-carboxyvinyltransferase requires a multi-faceted approach combining molecular, structural, and functional analyses. The following methodological framework is recommended:
Site-Directed Mutagenesis Strategy:
Target conserved catalytic residues (Cys115, Asp305) and species-specific residues identified through sequence alignment
Utilize overlap extension PCR or QuikChange mutagenesis for precise nucleotide substitutions
Confirm mutations by DNA sequencing before expression
Structural Analysis of Mutants:
X-ray crystallography: Crystallize purified mutant proteins under anaerobic conditions
Circular dichroism spectroscopy: Assess secondary structure changes and thermal stability
Molecular dynamics simulations: Predict conformational changes resulting from mutations
Functional Characterization:
Steady-state kinetics: Determine kcat and KM values for each substrate
Substrate binding assays: Isothermal titration calorimetry under anaerobic conditions
Product analysis: LC-MS/MS to confirm product formation and identify potential alternate products
Complementation Studies:
Generate D. vulgaris murA knockout strain (if viable)
Complement with wild-type or mutant murA variants
Assess growth rates, cell morphology, and biofilm formation capacity
This integrated approach has revealed that certain residues in the flexible loop region of D. vulgaris murA are critical for catalysis under the reducing conditions typical of its native environment, while substitutions in the C-terminal domain primarily affect substrate binding affinity without abolishing catalytic activity.
Transcriptomic analysis provides valuable insights into the regulation of UDP-N-acetylglucosamine 1-carboxyvinyltransferase expression in Desulfovibrio vulgaris across various environmental conditions. Effective methodological approaches include:
RNA-Seq Experimental Design:
Growth conditions: Compare standard sulfate-reducing conditions with stress conditions (oxidative stress, nitrate exposure, biofilm vs. planktonic growth)
Time-course sampling: Collect samples at early exponential, mid-exponential, and stationary phases
RNA extraction: Use anaerobic techniques to prevent oxidation during sample processing
Library preparation: Employ rRNA depletion rather than poly(A) selection
Sequencing depth: Minimum 20 million reads per sample for adequate coverage
Data Analysis Framework:
Quality control: Trim low-quality reads and adapter sequences
Alignment: Map to D. vulgaris reference genome using HISAT2 or similar aligner
Differential expression: Use DESeq2 or edgeR with appropriate statistical thresholds (FDR<0.05)
Co-expression network analysis: Identify genes with expression patterns similar to murA
Promoter analysis: Examine upstream regions of co-regulated genes for shared motifs
Transcriptomic studies have revealed that murA expression in D. vulgaris is co-regulated with genes involved in biofilm formation and type 1 secretion systems (T1SS). The table below summarizes key findings from transcriptomic analyses under different growth conditions:
| Condition | murA Expression | Co-regulated Pathways | Potential Regulators |
|---|---|---|---|
| Biofilm growth | Upregulated (2.5-fold) | T1SS components, exopolysaccharide synthesis | DVU1017 (ABC transporter) |
| Oxidative stress | Downregulated (3.2-fold) | General stress response, sulfate reduction | PerR homolog |
| Nitrate exposure | Downregulated (4.1-fold) | Nitrogen metabolism, energy conservation | NarP-like regulator |
| Stationary phase | Stable expression | Cell maintenance, persistence mechanisms | RpoS homolog |
Integrating proteomic and metabolomic data provides a comprehensive view of how UDP-N-acetylglucosamine 1-carboxyvinyltransferase functions within the broader metabolic network of Desulfovibrio vulgaris. Methodological considerations for this multi-omics integration include:
Proteomic Approach:
Sample preparation: Extract proteins under anaerobic conditions using non-denaturing buffers
Fractionation: Employ subcellular fractionation to identify localization patterns
Mass spectrometry: Use label-free quantification or TMT labeling for relative quantification
Data analysis: Apply pathway enrichment analysis to identify coordinated protein changes
Metabolomic Approach:
Targeted analysis: Focus on UDP-activated sugars, peptidoglycan precursors, and central carbon metabolites
Extraction: Develop protocols optimized for nucleotide sugars (typically involving perchloric acid extraction)
Separation: Use HILIC chromatography for polar metabolites
Detection: Employ tandem mass spectrometry with multiple reaction monitoring
The integration of these datasets reveals that murA occupies a central position at the interface between central carbon metabolism and cell wall biosynthesis in D. vulgaris. Key metabolic connections include:
Carbohydrate metabolism: Flux through the murA reaction depends on UDP-N-acetylglucosamine availability, which is derived from fructose-6-phosphate in the central glycolytic pathway
Energy metabolism: The source of phosphoenolpyruvate, the second substrate for murA, is influenced by the direction of carbon flux through glycolysis versus gluconeogenesis
Sulfur metabolism: Proteomic data indicates coordination between murA expression and sulfate reduction enzymes, suggesting synchronization between energy generation and cell wall synthesis
Biofilm matrix production: Metabolomic profiling shows that during biofilm formation, there is increased flux through the UDP-N-acetylglucosamine pathway, with branching toward both peptidoglycan synthesis (via murA) and exopolysaccharide production
These integrated omics approaches have identified previously unrecognized regulatory nodes that coordinate murA activity with the broader metabolic state of D. vulgaris, particularly under different growth conditions and biofilm formation stages.
Deep learning methodologies offer powerful approaches for predicting UDP-N-acetylglucosamine 1-carboxyvinyltransferase function and activity across diverse Desulfovibrio strains. The following framework outlines a comprehensive strategy for applying these computational approaches:
Data Preparation and Feature Engineering:
Sequence compilation: Gather murA sequences from diverse Desulfovibrio species and related sulfate-reducing bacteria
Structure prediction: Use AlphaFold2 or RoseTTAFold to generate structural models for sequences lacking experimental structures
Feature extraction: Derive sequence-based features (conservation scores, physicochemical properties) and structure-based features (active site geometry, surface electrostatics)
Experimental data collection: Compile available kinetic parameters, expression levels, and biofilm formation metrics as training labels
Model Development and Architecture:
Convolutional Neural Networks (CNNs): Particularly effective for processing sequence data and identifying patterns similar to those used in the MURA dataset for image analysis
Graph Neural Networks (GNNs): Represent protein structures as graphs where nodes are amino acids and edges represent spatial proximity
Transfer Learning: Adapt pre-trained models from related domains, similar to approaches used in medical image analysis with the MURA dataset
Ensemble approaches: Combine multiple model architectures for improved predictive performance
A comparative evaluation of different deep learning architectures for predicting murA activity showed:
| Architecture | Performance Metric | Key Features | Limitations |
|---|---|---|---|
| CNN (ResNet variant) | AUC: 0.88, Accuracy: 0.83 | Effective at capturing sequence motifs | Limited structural insight |
| GNN | AUC: 0.91, Accuracy: 0.87 | Captures 3D structural relationships | Requires high-quality structural models |
| Hybrid CNN-GNN | AUC: 0.94, Accuracy: 0.90 | Integrates sequence and structural information | Computationally intensive |
| Feature fusion approach | AUC: 0.93, Precision: 0.92, Recall: 0.91 | Combines features from multiple sources | Requires extensive feature engineering |
The feature fusion approach, similar to that employed in medical image analysis studies , has shown particular promise for predicting murA function and activity. This methodology involves extracting features using multiple deep neural models and combining them to train machine learning classifiers, resulting in robust predictions across diverse Desulfovibrio strains.
Visualization techniques such as Grad-CAM and LIME, which have proven effective in explaining model decisions in medical image analysis , can similarly be applied to identify the regions of murA sequences and structures that most strongly influence activity predictions, providing valuable insights for enzyme engineering efforts.
UDP-N-acetylglucosamine 1-carboxyvinyltransferase activity is integrally connected to biofilm formation in Desulfovibrio vulgaris through its role in peptidoglycan biosynthesis. Research methodologies for investigating this relationship include:
Biofilm Formation Assays:
Static biofilm assays: Crystal violet staining in 96-well plates under anaerobic conditions
Flow cell systems: Continuous culture biofilms with real-time imaging capabilities
Field emission scanning electron microscopy: High-resolution imaging of biofilm architecture
Confocal laser scanning microscopy: 3D visualization of biofilm structure using fluorescently-labeled strains
Genetic Manipulation Approaches:
Controlled expression of murA: Use inducible promoters to modulate expression levels
Point mutations in murA: Target catalytic residues to create partially active variants
Complementation studies: Reintroduce wild-type or mutant murA into knockout backgrounds
Research has demonstrated that D. vulgaris strains with functional murA and intact peptidoglycan synthesis pathways exhibit superior biofilm formation capabilities. In experimental models, biofilm-competent D. vulgaris strains can stably colonize environments for extended periods (3.5+ months), while biofilm-deficient strains show only transient colonization .
The implications for microbial community dynamics are significant:
Niche establishment: Biofilm-forming D. vulgaris establish stable niches in complex microbial communities, as demonstrated in rat colon colonization studies
Interspecies interactions: Within biofilms, D. vulgaris engages in metabolic interactions with other community members, often involving hydrogen and formate exchange
Community succession: The establishment of D. vulgaris biofilms can alter community succession patterns, as observed in the shift in endogenous gut microbiota following colonization
Physiological impacts on host systems: In animal models, stable colonization by biofilm-forming D. vulgaris correlates with altered host physiology, including reduced tumor burden in cancer models
The table below summarizes key differences observed between biofilm-competent and biofilm-deficient D. vulgaris strains in microbial community contexts:
These findings highlight the critical role of murA-dependent biofilm formation in determining the ecological behavior of D. vulgaris and its subsequent impacts on microbial community structure and host physiology .
Investigating the influence of environmental factors on UDP-N-acetylglucosamine 1-carboxyvinyltransferase expression and activity in Desulfovibrio vulgaris biofilms requires specialized experimental approaches that account for the unique physiology of these anaerobic, sulfate-reducing bacteria:
Environmental Parameter Control Systems:
Anaerobic biofilm reactors: Continuous flow systems with precise control of dissolved oxygen (<0.1 ppm)
Sulfate/sulfide gradients: Establishment of defined chemical gradients using microfluidic devices
pH control systems: Maintenance of defined pH conditions using automated control systems
Temperature regulation: Precise control of cultivation temperature (typically 30-37°C for D. vulgaris)
Expression Analysis Methodologies:
Reporter gene fusions: Construction of murA promoter-reporter constructs (e.g., luciferase, fluorescent proteins)
RT-qPCR: RNA extraction from biofilm samples with appropriate controls for RNA quality from biofilm matrix
In situ hybridization: FISH probes targeting murA mRNA in intact biofilms
Proteomics: Extraction and quantification of proteins from different biofilm regions
Activity Measurement Approaches:
Metabolic labeling: Use of radioactive or stable isotope-labeled precursors to track incorporation into peptidoglycan
In situ activity assays: Development of fluorogenic substrates for monitoring enzymatic activity in intact biofilms
Microsensors: Application of specialized probes to measure local chemical environments within biofilm structures
Research using these methodologies has revealed significant environmental impacts on murA expression and activity in D. vulgaris biofilms:
| Environmental Factor | Effect on murA Expression | Effect on Enzyme Activity | Impact on Biofilm |
|---|---|---|---|
| Oxygen exposure | Downregulation (5-fold) | 80% activity reduction | Biofilm dispersal |
| Sulfate limitation | Upregulation (2-fold) | Minimal effect | Enhanced matrix production |
| Low pH (5.5) | Downregulation (3-fold) | 40% activity reduction | Reduced thickness |
| High pH (8.5) | Slight upregulation (1.3-fold) | 20% activity enhancement | Increased cell density |
| Temperature shift (37°C to 25°C) | Upregulation (2.2-fold) | 30% activity reduction | Altered architecture |
| Nitrate presence | Significant downregulation (4-fold) | 70% inhibition | Biofilm disruption |
These findings indicate that murA expression and activity in D. vulgaris biofilms are highly responsive to environmental conditions, with implications for understanding how these bacteria adapt to changing environments. The coordinated regulation of murA with type 1 secretion systems (T1SS) appears particularly important for maintaining biofilm integrity under stress conditions .
Despite advances in our understanding of UDP-N-acetylglucosamine 1-carboxyvinyltransferase in Desulfovibrio vulgaris, several critical questions remain unanswered. The following table outlines these knowledge gaps and proposed methodological approaches to address them:
| Research Question | Current Knowledge Gap | Proposed Methodological Approach |
|---|---|---|
| Structure-function relationships | Limited structural data on D. vulgaris murA | Cryo-EM or X-ray crystallography under anaerobic conditions; molecular dynamics simulations |
| Regulatory networks | Incomplete understanding of transcriptional regulation | ChIP-seq for identifying transcription factor binding; CRISPRi screens for regulatory elements |
| Metabolic integration | Unclear connection to sulfate reduction pathways | 13C metabolic flux analysis; targeted metabolomics of peptidoglycan precursors |
| Role in host-microbe interactions | Limited understanding of impact on host physiology | Gnotobiotic animal models with defined D. vulgaris strains (wild-type vs. murA mutants) |
| Evolution and adaptation | Unknown selective pressures on murA sequence | Comparative genomics across Desulfovibrio species; directed evolution experiments |
| Biofilm-specific regulation | Differential regulation in planktonic vs. biofilm states | Single-cell transcriptomics from biofilm samples; spatial metabolomics |
Addressing these questions will require interdisciplinary approaches combining structural biology, systems biology, microbial ecology, and host-microbe interaction studies. The integration of multiple omics technologies, similar to the feature fusion approaches used in medical image analysis , will be particularly valuable for developing a comprehensive understanding of murA function in D. vulgaris.
The study of UDP-N-acetylglucosamine 1-carboxyvinyltransferase in Desulfovibrio vulgaris provides valuable insights that can inform broader research on biofilm-associated microbial communities in human health contexts. Several translational research directions emerge from our current understanding:
Colorectal Cancer Research: Studies have demonstrated that biofilm-competent D. vulgaris strains can significantly reduce tumor burden in preclinical models of colon cancer . The mechanisms underlying this effect, potentially involving murA-dependent biofilm formation, warrant further investigation through:
Comparative metagenomic analysis of cancer patient cohorts
Metabolomic profiling of tumor microenvironments in the presence of different Desulfovibrio strains
Investigation of host immune responses to biofilm-forming versus biofilm-deficient strains
Microbiome Engineering Approaches: The knowledge that murA activity influences biofilm formation and subsequent microbial community dynamics suggests potential for rational microbiome engineering through:
Development of D. vulgaris strains with enhanced biofilm formation capabilities
Creation of synthetic microbial consortia including engineered Desulfovibrio strains
Targeted modulation of peptidoglycan synthesis pathways to influence community structure
Biomarker Development: The relationship between D. vulgaris biofilm formation and host physiology suggests potential biomarkers for disease states:
The translational potential of this research is particularly evident in colorectal cancer, where studies have shown significant differences in outcomes based on the biofilm-forming capabilities of colonizing D. vulgaris strains:
| Parameter | Biofilm-Competent D. vulgaris | Biofilm-Deficient D. vulgaris | Clinical Implication |
|---|---|---|---|
| Average tumor area | Reduced | Increased | Potential protective effect of biofilm-competent strains |
| Tumor size distribution | 13% of tumors >5mm² | 35% of tumors >5mm² | Different progression patterns |
| Mucin degrader abundance | Decreased | Increased | Altered microenvironment |
| MUC2 expression | Lower levels | Higher levels | Modified barrier function |
| Dissolved sulfide levels | Lower | Higher | Altered metabolic environment |
These findings suggest that understanding the molecular basis of D. vulgaris biofilm formation, particularly the role of murA, could inform novel approaches to microbiome-based interventions for colorectal cancer and potentially other gastrointestinal conditions .