Gamma-glutamyl phosphate reductase (proA) is an enzyme that plays a crucial role in the biosynthesis of proline, an amino acid vital for protein synthesis, osmotic stress tolerance, and various other cellular functions . Specifically, proA catalyzes the NADPH-dependent reduction of L-glutamate 5-phosphate into L-glutamate 5-semialdehyde and phosphate, which is the second step in proline biosynthesis from glutamate .
ProA functions as a reductase, utilizing NADPH to reduce L-glutamate 5-phosphate, also known as gamma-L-glutamyl 5-phosphate (GP), into L-glutamate 5-semialdehyde .
The reaction catalyzed by proA can be represented as follows:
L-glutamate 5-phosphate + NADPH + H+ → L-glutamate 5-semialdehyde + NADP+ + phosphate
This step is essential in the proline biosynthesis pathway, and the activity of proA is critical for maintaining proline levels within the cell .
Proline is an essential amino acid with diverse roles in bacteria:
Protein Structure: Proline's unique cyclic structure imparts rigidity to polypeptide chains, influencing protein folding and stability.
Osmoprotection: Proline acts as an osmoprotectant, helping cells maintain osmotic balance under stress conditions.
Stress Tolerance: Proline accumulation is a common response to environmental stresses, such as drought, salinity, and temperature extremes.
Virulence: ProA influences the expression of T3SS genes and host invasion in Ralstonia solanacearum .
In Ralstonia solanacearum, a bacterial plant pathogen, proA has been identified as a key enzyme involved in proline biosynthesis . Studies have shown that proA mutants are proline auxotrophs, unable to grow in minimal medium unless supplemented with proline . This confirms that ProA is responsible for proline biosynthesis from glutamate in this bacterium .
Furthermore, ProA in R. solanacearum has been found to play roles beyond proline biosynthesis, particularly in the regulation of the type three secretion system (T3SS), which is crucial for pathogenicity . Deletion of proA impairs T3SS expression, suggesting that ProA is involved in the mechanisms that control the T3SS genes .
Note: While we prioritize shipping the format currently in stock, please specify any format requirements in your order notes for customized preparation.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
KEGG: dvu:DVU1953
STRING: 882.DVU1953
ProA in Desulfovibrio vulgaris likely functions similarly to other bacterial species, catalyzing the second reaction in the proline biosynthesis pathway by converting gamma-glutamyl phosphate to glutamate-5-semialdehyde. This reaction is essential for proline formation from glutamate, as demonstrated in related bacteria such as Ralstonia solanacearum. In R. solanacearum, ProA mutants are proline auxotrophs that fail to grow in minimal media, with growth restored only through proline supplementation rather than glutamate addition . Although specific details for D. vulgaris are not fully characterized in the provided research, the enzyme likely plays a similar fundamental role in amino acid metabolism.
Desulfovibrio vulgaris Hildenborough has a complex genome with multiple regulatory systems. While specific information about ProA's genetic organization is limited in the provided research, we know that D. vulgaris contains numerous enhancer binding proteins (EBPs), more than most sequenced bacteria . This suggests sophisticated regulatory control over various metabolic pathways, potentially including the proline biosynthesis pathway where ProA functions. Like other bacterial species, the proA gene in D. vulgaris likely exists as part of an operon structure involved in amino acid biosynthesis, though its specific genomic context would require targeted sequence analysis.
For recombinant production of D. vulgaris proteins, researchers typically employ E. coli-based expression systems using vectors with inducible promoters. Based on methodologies used for similar proteins, successful expression often requires optimization of codon usage, as the GC content of D. vulgaris differs from expression hosts. Expression constructs should include affinity tags (such as His-tag or GST) for purification, and expression conditions must be optimized to prevent inclusion body formation. For anaerobic proteins like those from D. vulgaris, expression under microaerobic or anaerobic conditions may improve protein folding and activity. Temperature reduction during induction (to 16-25°C) and specialized E. coli strains designed for expression of difficult proteins could enhance production of soluble, active enzyme.
The relationship between ProA and biofilm formation in D. vulgaris presents an intriguing research area. In D. vulgaris Hildenborough, biofilm formation is critical for host colonization, as demonstrated in rat colon studies where biofilm-competent strains could stably colonize for up to 3.5 months . While direct evidence linking ProA to biofilm formation in D. vulgaris is not explicitly stated in the research, we can draw parallels from other bacterial systems. For instance, in R. solanacearum, ProA has roles beyond proline biosynthesis, affecting pathogenicity mechanisms .
In D. vulgaris, type 1 secretion systems (T1SS) are essential for biofilm formation and colonization of the rat colon . Since amino acid metabolism can influence bacterial surface properties and secretion systems, ProA may contribute to biofilm formation through:
Providing proline for extracellular matrix proteins
Influencing cell surface hydrophobicity through membrane protein modifications
Affecting signaling pathways that regulate biofilm development
Researchers investigating this relationship should consider gene knockout studies coupled with biofilm quantification assays and colonization experiments to elucidate ProA's specific contributions.
The structural and functional analysis of D. vulgaris ProA compared to homologs in pathogenic bacteria reveals important evolutionary adaptations. While detailed structural information for D. vulgaris ProA is not provided in the search results, comparative analysis would typically examine:
Catalytic domain conservation: The core catalytic domain of gamma-glutamyl phosphate reductases is generally conserved across species, but D. vulgaris ProA may contain modifications reflecting its adaptation to anaerobic environments.
Substrate binding pocket variations: Subtle differences in the substrate binding pocket could affect substrate specificity and catalytic efficiency.
Regulatory domains: Unlike pathogenic bacteria where ProA may have additional regulatory functions (as seen in R. solanacearum where ProA affects type three secretion system expression ), D. vulgaris ProA might have specialized domains related to its role in sulfate-reducing metabolism.
Protein-protein interaction surfaces: Differences in interaction surfaces could reflect species-specific metabolic integration.
Researchers should employ molecular modeling, site-directed mutagenesis, and enzyme kinetics studies to characterize these differences comprehensively.
The interplay between sulfide production and ProA activity in D. vulgaris presents a complex relationship with significant implications for colonization and disease processes. D. vulgaris, as a sulfate-reducing bacterium, produces sulfide as a metabolic byproduct. Research indicates that sulfide levels correlate with adenomagenesis in rat models, with higher sulfide levels associated with increased adenoma burden .
The relationship with ProA might involve several mechanisms:
Metabolic coupling: ProA's role in proline biosynthesis may be metabolically linked to sulfate reduction pathways, affecting energy availability for sulfide production.
Redox balance: Both pathways involve redox reactions that could influence each other through shared electron carriers or redox-sensitive regulatory mechanisms.
Colonization effects: Since both biofilm formation (potentially influenced by ProA) and sulfide production affect colonization capabilities, there may be regulatory crosstalk between these systems.
Interestingly, biofilm-competent D. vulgaris strains that successfully colonized rat colons were associated with reduced dissolved sulfide levels in feces and reduced adenoma burden . This suggests a possible protective effect that warrants investigation into whether ProA activity influences this relationship.
Optimizing assay conditions for recombinant D. vulgaris ProA enzymatic activity requires careful consideration of this anaerobic organism's biochemical requirements. Based on similar enzyme systems, the following conditions would likely be optimal:
Buffer composition: HEPES or Tris buffer (50-100 mM) at pH 7.5-8.0 with 50-150 mM NaCl to maintain ionic strength.
Reducing environment: Inclusion of reducing agents (1-5 mM DTT or 0.1-1 mM TCEP) to maintain the enzyme's redox-sensitive residues in reduced state.
Anaerobic conditions: As D. vulgaris is an anaerobic organism, its enzymes often perform optimally under anaerobic or microaerobic conditions. Enzyme assays should be conducted in an anaerobic chamber or using oxygen-scavenging systems.
Divalent cation requirement: Addition of Mg²⁺ or Mn²⁺ (1-5 mM) as potential cofactors.
Temperature and kinetics: Assays typically performed at 30-37°C with reaction times determined through preliminary time-course experiments.
The assay itself would likely involve monitoring the conversion of gamma-glutamyl phosphate to glutamate-5-semialdehyde, which could be quantified using chromatographic methods coupled with appropriate detection systems. A coupled enzymatic assay that generates a spectrophotometrically detectable product could also be developed for higher throughput analysis.
Generating stable ProA knockouts in D. vulgaris requires specialized approaches due to the challenging nature of genetic manipulation in this anaerobic bacterium. Based on successful approaches with similar organisms, the following strategies are recommended:
Homologous recombination-based methods: Using plasmid systems similar to the pK18mobsacB-based approach, where DNA fragments flanking the proA gene are conjugated and subcloned into an appropriate vector . This approach has been successful in related bacteria.
Suicide vector delivery: Conjugation using E. coli S17-1 as a donor strain to transfer the knockout construct into D. vulgaris .
Marker selection: Implementing appropriate antibiotic resistance markers that function in D. vulgaris, followed by sacB-based counter-selection for identifying double crossover events.
Verification strategies: Confirmation through colony PCR with appropriate primer pairs and sequencing to ensure in-frame deletion without polar effects on downstream genes .
Complementation system: Establishing a reliable complementation system such as the Tn7-based site-specific chromosomal integration system, which has been effective in restoring phenotypes in other bacteria .
For functional validation, growth assays in minimal media with and without proline supplementation should be conducted to confirm the auxotrophic phenotype expected from ProA deficiency, as demonstrated with ProA mutants in R. solanacearum .
Differentiating between direct and indirect effects of ProA mutation on biofilm formation requires a multi-faceted experimental approach. Researchers should implement these strategies:
Biochemical complementation: Compare biofilm formation in ProA mutants with exogenous proline supplementation versus genetic complementation with the intact proA gene. If proline supplementation alone doesn't restore normal biofilm formation (as seen with T3SS expression in R. solanacearum ), this suggests ProA has functions beyond proline biosynthesis.
Temporal gene expression analysis: Perform time-course RNA-seq experiments comparing wild-type and ProA mutants during biofilm development stages to identify differentially expressed genes, particularly those involved in biofilm formation.
Protein interaction studies: Conduct pull-down assays or bacterial two-hybrid screens to identify proteins that directly interact with ProA, potentially revealing non-catalytic functions.
Metabolomic analysis: Compare metabolite profiles between wild-type and ProA mutants to identify metabolic shifts beyond proline depletion that might affect biofilm formation.
Structural mutant analysis: Generate mutations that affect ProA's catalytic activity versus potential protein-protein interaction domains to separate enzymatic and non-enzymatic functions.
By integrating these approaches, researchers can build a comprehensive understanding of ProA's direct contributions to biofilm formation versus indirect effects stemming from metabolic disruption.
When analyzing colonization dynamics of wild-type versus ProA-mutant D. vulgaris in animal models, researchers should employ robust statistical methods that account for the temporal and spatial complexity of bacterial colonization. Based on successful approaches in similar studies, the following statistical framework is recommended:
Longitudinal data analysis: Mixed-effects models for repeated measures data that can account for individual animal variation over time. This is particularly important when tracking colonization over extended periods, such as the 3.5-month colonization observed with biofilm-competent D. vulgaris strains .
Survival analysis techniques: Kaplan-Meier curves and Cox proportional hazards models to analyze time-to-colonization or persistence duration differences between wild-type and mutant strains.
Microbial community analysis: When examining effects on the host microbiome, approaches like PERMANOVA or ANOSIM should be used to assess community composition differences, similar to the analysis of microbiome shifts observed in the rat studies .
Spatial statistics: For tissue-specific colonization analysis, spatial statistical methods should be applied to differentiate colonization patterns between different gut compartments.
Power analysis considerations: A priori power calculations based on preliminary data or similar studies are essential, with typical animal studies requiring 8-12 animals per group to detect biologically meaningful differences in colonization.
When presenting results, visualization through longitudinal curves with confidence intervals, stacked bar charts for community composition, and heat maps for spatial distribution provides the most informative representation of colonization dynamics.
Reconciling contradictory findings regarding D. vulgaris and sulfide production in colorectal cancer studies requires careful consideration of methodological differences, contextual factors, and biological complexity. The literature presents seemingly contradictory observations where Desulfovibrio spp. have been associated with both healthy controls and disease states in colorectal cancer studies .
To address these contradictions, researchers should:
Context-dependent analysis: Consider the biofilm competency of different strains, as biofilm-competent D. vulgaris strains were associated with reduced adenoma burden in rat models . This suggests functional variations among strains may explain divergent outcomes.
Spatial arrangement considerations: Investigate the spatial arrangement of the gut microbiome, as the research suggests "sulfide has different effects contingent on the spatial arrangement of the GM [gut microbiota]" . Different sampling methods (fecal vs. mucosal biopsies) may capture different microbial populations.
Multi-omics integration: Combine microbiome data with metabolomics, transcriptomics, and host genetic information to develop models that account for host-microbe interactions. This approach has revealed that sulfide levels correlate with expression of genes like MUC2 and DNA damage response genes .
Dose-response relationships: Establish dose-response curves for sulfide production and disease outcomes, as concentration thresholds may explain contradictory effects.
Temporal dynamics: Consider the timing of colonization and sulfide production relative to disease initiation and progression, as effects may differ during different stages of carcinogenesis.
By systematically addressing these factors, researchers can develop more nuanced models that explain the seemingly contradictory roles of D. vulgaris and sulfide production in colorectal cancer.
Investigating non-canonical functions of ProA in D. vulgaris presents exciting research opportunities. Drawing parallels from findings in R. solanacearum, where ProA has functions beyond proline biosynthesis affecting T3SS expression even under proline-supplemented conditions , researchers should consider these approaches:
Comprehensive phenotypic profiling: Compare wild-type and ProA-deficient strains under various stresses (oxidative, osmotic, pH) even with proline supplementation to identify proline-independent phenotypes.
Interactome mapping: Employ techniques like BioID or affinity purification coupled with mass spectrometry to identify proteins that interact with ProA, potentially revealing unexpected biological pathways.
Domain-specific mutagenesis: Generate mutations that affect specific ProA domains while preserving catalytic activity to dissect non-catalytic functions.
Transcriptomic analysis under stress conditions: Compare gene expression profiles between wild-type and ProA mutants under conditions relevant to colonization or biofilm formation, even with proline supplementation.
Heterologous expression studies: Express D. vulgaris ProA in other bacterial species and assess effects on their phenotypes to identify conserved non-canonical functions.
These approaches would help elucidate whether ProA in D. vulgaris, like its counterpart in R. solanacearum, has evolved additional regulatory or structural roles beyond its enzymatic function in proline biosynthesis.
The expression and function of ProA in D. vulgaris likely undergoes complex regulation during host colonization in response to changing environmental conditions. Research strategies to investigate this should consider:
In vivo expression profiling: Using reporter gene fusions or RNA-seq analysis of D. vulgaris recovered from different host compartments to track ProA expression during colonization. This approach could reveal whether ProA expression varies between different niches within the gut, similar to how biofilm-forming D. vulgaris showed differential colonization patterns in rat studies .
Environmental response elements: Analyzing the promoter region of the proA gene for binding sites of environmental response regulators that might control expression under different conditions.
Post-translational modification analysis: Investigating whether ProA undergoes post-translational modifications in response to environmental cues that might alter its activity or interaction partners.
Nutrient availability effects: Exploring how varying levels of amino acids, particularly proline and glutamate, in different host compartments might affect ProA expression through feedback mechanisms.
Host immune factor interactions: Determining whether host defense molecules like antimicrobial peptides or reactive oxygen species alter ProA expression or activity, potentially as part of a bacterial stress response.
Understanding these environmental response mechanisms would provide insights into how D. vulgaris adapts its proline metabolism and potential ProA-mediated biofilm formation during the dynamic process of host colonization.