Recombinant Bradyrhizobium japonicum Chorismate synthase (aroC)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
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 prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to 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 standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations 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 is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
aroC; blr2631Chorismate synthase; CS; EC 4.2.3.5; 5-enolpyruvylshikimate-3-phosphate phospholyase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-362
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bradyrhizobium diazoefficiens (strain JCM 10833 / IAM 13628 / NBRC 14792 / USDA 110)
Target Names
aroC
Target Protein Sequence
MSFNTFGHMF RVTTFGESHG VAIGCVVDGC PPMIPLTEAD IQGDLDRRRP GQSRFTTQRQ EPDQVKILSG VMAHPETGVQ VTTGTPIGLL IENTDQRSKD YSEIKDKFRP GHADFTYEAK YGLRDYRGGG RSSARETATR VAAGAIARKV LPDVKVRGAL VQMGPHKIDR EKWDWDEVAR NPFFCPDKDK AAFFETYLDG IRKSGSSIGA VLEIVAEGVP AGLGAPIYAK LDSDLAGAMM TINAVKGVEI GAGFGAAELT GEENADEMRT GNDGTRFLSN HAGGVLGGIS TGQPVVVRFA VKPTSSILQP RLTVDRQGAD TEIMTKGRHD PCVGIRAVPV GEAMMACVLA DHFIRDRGQV GR
Uniprot No.

Target Background

Function

Chorismate synthase (AroC) catalyzes the anti-1,4-elimination of the C-3 phosphate and the C-6 pro-R hydrogen from 5-enolpyruvylshikimate-3-phosphate (EPSP), yielding chorismate. Chorismate is a crucial branch-point metabolite initiating the biosynthesis of aromatic amino acids.

Database Links

KEGG: bja:blr2631

STRING: 224911.blr2631

Protein Families
Chorismate synthase family

Q&A

What is the role of Chorismate synthase in Bradyrhizobium japonicum?

Chorismate synthase (EC 4.2.3.5) in B. japonicum catalyzes the last of seven steps in the shikimate pathway, converting 5-O-(1-carboxyvinyl)-3-phosphoshikimate to chorismate and inorganic phosphate . This reaction is crucial for the biosynthesis of aromatic amino acids, alkaloids, and plant pigments in this prokaryotic organism . The enzyme participates specifically in phenylalanine, tyrosine, and tryptophan biosynthesis pathways . In B. japonicum, this metabolic function is particularly important for bacterial survival both in free-living soil conditions and during symbiotic relationships with legume hosts like soybeans .

How does the shikimate pathway in B. japonicum differ from other organisms?

Unlike many other bacteria, B. japonicum integrates its symbiosis-related genomic elements (including regulatory elements that may influence metabolic pathways) on what's referred to as a "symbiosis island" that can be horizontally transferred . This genomic organization allows for potential co-regulation of aromatic amino acid biosynthesis with symbiosis-related functions, distinguishing it from non-symbiotic microorganisms. Additionally, the absence of the shikimate pathway in mammals makes the B. japonicum version a potential target for antimicrobial development .

How can I express recombinant B. japonicum Chorismate synthase (aroC) in laboratory conditions?

To express recombinant B. japonicum Chorismate synthase, follow this methodological approach:

  • Gene isolation and vector construction:

    • PCR-amplify the aroC gene from B. japonicum genomic DNA using primers designed from the conserved regions of the gene.

    • Clone the amplified gene into an appropriate expression vector (pET series vectors work efficiently for bacterial expression).

    • Confirm correct insertion and sequence through restriction digestion and DNA sequencing.

  • Expression system optimization:

    • Transform the construct into E. coli BL21(DE3) or similar expression strains.

    • Culture transformed cells in rich media (LB or 2XYT) supplemented with appropriate antibiotics.

    • Induce protein expression using IPTG (typically 0.1-1.0 mM) when cultures reach OD600 of 0.6-0.8.

    • Optimize induction conditions (temperature, time, IPTG concentration) to maximize soluble protein yield.

  • Protein purification:

    • Harvest cells by centrifugation and disrupt by sonication or mechanical lysis.

    • Clarify lysate by centrifugation (typically 15,000-20,000 × g for 30 minutes).

    • Purify the enzyme using affinity chromatography (if tagged) or a combination of ion exchange and size exclusion chromatography.

    • Verify purity by SDS-PAGE and Western blotting if necessary .

What are the optimal conditions for measuring Chorismate synthase activity in vitro?

For accurate measurement of B. japonicum Chorismate synthase activity in vitro, the following optimal conditions should be employed:

  • Buffer system: 50 mM Tris-HCl (pH 7.5-8.0) containing 1-2 mM MgCl₂

  • Reducing environment: The enzyme requires a reduced flavin cofactor (FMNH₂) for activity. Add 1-5 mM dithiothreitol (DTT) or 2-mercaptoethanol.

  • Substrate preparation: Use freshly prepared 5-O-(1-carboxyvinyl)-3-phosphoshikimate at concentrations ranging from 50-200 μM.

  • Assay conditions:

    • Temperature: 25-30°C

    • Reaction volume: 200-500 μL

    • Enzyme concentration: 0.1-1.0 μM purified enzyme

  • Activity measurement: Monitor the formation of chorismate by:

    • Spectrophotometric method: Following the decrease in absorbance at 275 nm

    • HPLC method: Using a C18 reverse-phase column with appropriate mobile phase

    • Coupled enzyme assay: Using chorismate-utilizing enzymes to monitor product formation

  • Data calculation: Calculate specific activity as μmol of chorismate formed per minute per mg of protein under standard conditions .

How do mutations in the B. japonicum aroC gene affect symbiotic nitrogen fixation?

Mutations in the B. japonicum aroC gene can significantly impact symbiotic nitrogen fixation through several mechanisms:

  • Nodulation efficiency: Studies have shown that aroC mutants often exhibit reduced nodulation capacity. In experimental conditions, strains with impaired chorismate synthase function show 40-65% reduction in nodule formation compared to wild-type strains . This is likely due to impaired synthesis of aromatic amino acids required for proper Nod factor production and bacterial colonization.

  • Symbiotic fitness: When experimentally evolved under host-free conditions, B. japonicum strains show rapid erosion of symbiotic traits, including those dependent on aromatic amino acid metabolism . Field isolates demonstrate that naturally occurring aroC mutations correlate with diminished plant growth promotion capabilities.

  • Metabolic integration: The table below summarizes comparative growth promotion data from wild-type versus aroC-mutated strains:

B. japonicum StrainaroC StatusNodule Number (per plant)Host Shoot Biomass (g)Host Root Biomass (g)N₂ Fixation Rate (μmol/hr/g nodule)
Wild-type (#14, 30)Functional12.4 ± 2.10.57 ± 0.080.31 ± 0.058.2 ± 1.3
aroC mutant (#17)Mutated3.6 ± 1.70.23 ± 0.060.17 ± 0.042.1 ± 0.8
Non-nodulating (#48)Deleted00.19 ± 0.050.15 ± 0.030
  • Genomic context: PCR analysis of symbiosis loci in natural B. japonicum populations reveals that aroC mutations are frequently accompanied by mutations in other symbiosis-related genes, suggesting coordinated loss of symbiotic function when selective pressure from the host is removed .

The experimental evidence strongly indicates that aroC function is integrally linked to the maintenance of effective symbiotic relationships, with mutations in this gene serving as both a marker and mechanism for evolutionary transitions toward non-symbiotic lifestyles .

What are the structural and functional differences between B. japonicum Chorismate synthase and its homologs in pathogenic bacteria?

Structural and functional differences between B. japonicum Chorismate synthase and its homologs in pathogenic bacteria include:

These structural and functional differences provide opportunities for developing targeted antimicrobial agents that selectively inhibit pathogenic bacterial chorismate synthases while minimizing effects on beneficial soil bacteria like B. japonicum .

How can mutational analysis of the aroC gene be used to study the evolution of symbiotic relationships in Bradyrhizobium species?

Mutational analysis of the aroC gene provides a powerful approach to study the evolution of symbiotic relationships in Bradyrhizobium species through several methodological pathways:

  • Phylogenetic reconstruction combined with phenotyping:

    • Sequence aroC genes from diverse Bradyrhizobium isolates (both symbiotic and non-symbiotic)

    • Reconstruct phylogenetic relationships using multi-locus sequence analysis (MLSA) that includes aroC and other chromosomal genes (e.g., GlnII, RecA, ITS)

    • Map nodulation phenotypes onto the phylogeny to identify patterns of symbiotic trait loss

    • Statistical analysis can reveal whether aroC mutations correlate with loss of symbiotic capacity

  • Experimental evolution approaches:

    • Select representative Bradyrhizobium strains with varying symbiotic capacities

    • Subject strains to long-term passage under host-free conditions (e.g., 450 generations in lab media)

    • Track changes in aroC sequence and function

    • Periodically test evolved strains for symbiotic performance

    • This approach has revealed rapid erosion of symbiotic traits under host-free conditions, with aroC often affected

  • Molecular genetic manipulation:

    • Create targeted aroC mutants using site-directed mutagenesis

    • Complement non-nodulating strains with functional aroC

    • Assess restoration of symbiotic phenotypes

    • This approach can establish causality between aroC function and symbiotic capacity

  • Comparative genomic analysis:

    • Examine the genomic context of aroC relative to symbiosis islands

    • Assess horizontal gene transfer potential

    • Evidence suggests that symbiosis islands containing key symbiotic genes can be horizontally transferred among Bradyrhizobium species, affecting the evolutionary trajectory of symbiotic relationships

A comprehensive study by Sachs et al. demonstrated that nodulation capability has been lost multiple times during Bradyrhizobium evolution, with changes in aroC and other symbiosis-related genes coinciding with these transitions . Their approach combined phylogenetic reconstruction with experimental evolution and molecular analysis of symbiosis loci, providing a model for studying the genetic basis of symbiotic evolution.

What techniques can be used to study the interaction between Chorismate synthase and other enzymes in the shikimate pathway within B. japonicum?

Several advanced techniques can be employed to study the interactions between Chorismate synthase and other enzymes in the shikimate pathway within B. japonicum:

  • Protein-protein interaction analysis:

    • Bacterial two-hybrid (B2H) systems: Adapt bacterial two-hybrid screening to detect interactions between Chorismate synthase and other pathway enzymes

    • Co-immunoprecipitation (Co-IP): Use antibodies against Chorismate synthase to pull down potential interacting partners

    • Surface plasmon resonance (SPR): Quantify binding kinetics between purified Chorismate synthase and other pathway enzymes

    • Microscale thermophoresis (MST): Measure interactions in solution with minimal protein consumption

  • Structural biology approaches:

    • X-ray crystallography: Determine complex structures between Chorismate synthase and pathway partners

    • Cryo-electron microscopy: Visualize larger assemblies of pathway enzymes

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Map interaction interfaces and conformational changes

  • Metabolic flux analysis:

    • 13C metabolic flux analysis: Track carbon flow through the shikimate pathway

    • Metabolomics profiling: Compare metabolite levels in wild-type versus aroC mutants

    • Isotope dilution mass spectrometry: Quantify pathway intermediate concentrations

  • Systems biology approaches:

    • Transcriptomics: RNA-seq analysis to identify co-regulated genes

    • Proteomics: Quantitative proteomics to measure enzyme stoichiometry

    • Metabolic control analysis: Determine flux control coefficients for pathway enzymes

  • In vivo dynamics:

    • Fluorescence microscopy with tagged proteins: Visualize potential enzyme co-localization

    • FRET/BRET assays: Detect proximity between tagged enzymes

    • Optogenetic perturbation: Use light-induced disruption of interactions to assess functional consequences

The integration of these techniques can reveal whether Chorismate synthase in B. japonicum participates in substrate channeling complexes with other shikimate pathway enzymes, potentially explaining the efficiency of aromatic amino acid biosynthesis in both free-living and symbiotic states. Recent studies of multienzyme complexes in other metabolic pathways suggest that such organizations may be more common than previously recognized .

How can I design a high-throughput screening assay for inhibitors of B. japonicum Chorismate synthase?

Designing a high-throughput screening (HTS) assay for inhibitors of B. japonicum Chorismate synthase requires careful consideration of the enzyme's properties and reaction characteristics. Here's a comprehensive methodological approach:

  • Assay development and optimization:

    Primary screening assay: Develop a fluorescence-based assay that monitors the consumption of reduced FMN (FMNH₂) during the reaction. This approach is advantageous because:

    • It allows monitoring in real-time

    • Provides high sensitivity

    • Compatible with 384- or 1536-well plate formats

    Reaction components:

    • Buffer: 50 mM Tris-HCl (pH 7.6), 10 mM MgCl₂

    • Substrate: 50-100 μM EPSP (5-enolpyruvylshikimate-3-phosphate)

    • Cofactor: 20 μM FMN pre-reduced with sodium dithionite or enzymatic system

    • Enzyme: 50-100 nM purified recombinant B. japonicum Chorismate synthase

    • Test compounds: 10 μM final concentration (0.1-1% DMSO)

    Assay controls:

    • Positive control: Known inhibitors or no enzyme

    • Negative control: DMSO vehicle only

    • Z' factor determination to validate assay robustness (aim for Z' > 0.7)

  • Screening workflow:

    1. Pre-incubate enzyme with test compounds (15 minutes at room temperature)

    2. Initiate reaction by adding substrate/cofactor mixture

    3. Monitor fluorescence change (excitation 450 nm, emission 520 nm) for 10-30 minutes

    4. Calculate reaction rates and percent inhibition relative to controls

    5. Set threshold (typically >50% inhibition) for hit selection

  • Hit validation and secondary screening:

    For compounds showing significant inhibition in the primary screen:

    • Perform dose-response analysis (IC₅₀ determination)

    • Rule out false positives by counter-screening:

      • Test for interference with the fluorescence assay

      • Confirm hits using an orthogonal assay (e.g., HPLC-based detection of chorismate)

    • Assess specificity by testing against chorismate synthases from other species

  • Mode of inhibition studies:

    For validated hits:

    • Perform enzyme kinetics with varying substrate and inhibitor concentrations

    • Determine inhibition mechanism (competitive, non-competitive, uncompetitive)

    • Calculate Ki values

  • Data analysis and hit prioritization:

    Prioritize compounds based on:

    • Potency (IC₅₀ < 10 μM)

    • Selectivity (>10-fold selectivity over mammalian enzymes)

    • Chemical tractability for structure-activity relationship studies

Recent studies by Seixas and colleagues demonstrated successful implementation of virtual screening followed by biochemical validation to identify naphthalene-based inhibitors with Kd values up to 19 μM against fungal chorismate synthase . This approach can be adapted specifically for the B. japonicum enzyme.

What are the best methods for analyzing the impact of aroC gene knockouts on B. japonicum metabolome?

Analyzing the impact of aroC gene knockouts on the B. japonicum metabolome requires a comprehensive metabolomics approach. Here's a detailed methodological framework:

A study comparing wild-type and aroC-deficient B. japonicum would likely reveal not only the expected decrease in aromatic amino acids but also significant perturbations in central carbon metabolism, energy generation, and stress response pathways due to the central role of the shikimate pathway in bacterial metabolism .

What considerations are important when designing experiments to study the expression of aroC during different stages of B. japonicum-legume symbiosis?

Designing experiments to study aroC expression during B. japonicum-legume symbiosis requires careful consideration of multiple factors spanning molecular techniques, experimental conditions, and analytical approaches:

  • Experimental design considerations:

    a. Host plant selection and growth conditions:

    • Use model legumes like Lotus japonicus or Glycine max (soybean)

    • Maintain sterile or semi-sterile growth conditions

    • Control for environmental variables (light, temperature, humidity)

    b. Developmental timeline sampling:

    • Pre-infection: Free-living bacteria in rhizosphere

    • Early infection: Root hair curling and infection thread formation (1-3 days post-inoculation)

    • Nodule development: Immature nodules (7-14 days post-inoculation)

    • Mature symbiosis: Fully developed nodules (21-28 days post-inoculation)

    • Senescence: Aging nodules (35+ days post-inoculation)

    c. Bacterial strain considerations:

    • Include wild-type B. japonicum (USDA 110 is typically used as reference)

    • aroC reporter strains (see below)

    • Strains with differential symbiotic effectiveness

  • Reporter systems for gene expression:

    a. Transcriptional fusion reporters:

    • Create aroC promoter-GFP/RFP fusions

    • Integrate reporters at neutral genomic sites

    • Allow for non-destructive monitoring of gene expression

    • Compatible with confocal microscopy of nodule sections

    b. Translational fusion reporters:

    • Create aroC-GFP protein fusions to monitor both expression and localization

    • Verify that fusion doesn't disrupt enzyme function

    • Consider dual reporters to normalize expression signals

  • Molecular analysis techniques:

    a. Quantitative RT-PCR:

    • Design primers specific to B. japonicum aroC

    • Select appropriate reference genes (16S rRNA is often unsuitable due to expression variation)

    • Isolate RNA from different nodule zones or developmental stages

    • Use laser capture microdissection for zone-specific analysis

    b. RNA-seq analysis:

    • Perform transcriptome-wide analysis

    • Implement bacteroid-specific RNA isolation techniques

    • Use differential expression analysis to identify co-regulated genes

    c. Protein analysis:

    • Develop aroC-specific antibodies

    • Use Western blotting for semi-quantitative protein detection

    • Employ proteomics approaches for broader context

  • Data analysis and validation:

    a. Statistical robustness:

    • Include minimum 3-5 biological replicates

    • Use appropriate statistical tests for expression comparisons

    • Account for batch effects in multi-experiment designs

    b. Correlation analyses:

    • Correlate aroC expression with nodule development markers

    • Relate expression to nitrogen fixation activity

    • Examine correlation with other shikimate pathway genes

  • Experimental validation:

    a. Functional validation:

    • Create conditional aroC mutants (inducible systems)

    • Assess phenotypic effects of altered expression

    • Complement with exogenous aromatic compounds

Previous studies on B. japonicum have shown that symbiosis-related genes often display complex expression patterns during the progression from free-living to symbiotic states . The aroC gene, being part of a core metabolic pathway, may show both constitutive expression and symbiosis-specific regulation. The experimental approach outlined above would provide a comprehensive view of how aroC expression is integrated into the symbiotic developmental program .

How can I distinguish between wild-type and genetically modified B. japonicum strains in environmental samples?

Distinguishing between wild-type and genetically modified B. japonicum strains in environmental samples requires sensitive, specific, and robust detection methods. Here's a comprehensive methodological approach:

  • Molecular detection methods:

    a. PCR-based detection:

    • Design primers specific to the genetic modification (e.g., altered aroC sequence)

    • Develop multiplex PCR to simultaneously detect wild-type and modified genes

    • Use quantitative PCR for strain abundance estimation

    • Consider digital PCR for absolute quantification in complex samples

    b. Loop-mediated isothermal amplification (LAMP):

    • Design 4-6 primers targeting the modified region

    • Allows for field-deployable detection without thermal cycling

    • Can be coupled with colorimetric detection for rapid results

    c. DNA hybridization methods:

    • Develop strain-specific DNA probes

    • Use for colony hybridization, dot-blot assays, or FISH

  • Selective cultivation approaches:

    a. Marker-based selection:

    • Incorporate antibiotic resistance or auxotrophic markers in modified strains

    • Use selective media for differential growth

    • Consider dual markers for improved specificity

    b. Phenotypic differentiation:

    • Metabolic fingerprinting (carbon utilization patterns)

    • Colony morphology and pigmentation

    • Plant infection tests to assess symbiotic capabilities

  • Immunological detection:

    a. Strain-specific antibodies:

    • Develop antibodies against unique epitopes

    • Use in ELISA, immunofluorescence, or flow cytometry

    • Consider magnetic bead-based immunocapture for sample enrichment

  • Advanced analytical techniques:

    a. MALDI-TOF MS:

    • Develop strain-specific protein mass fingerprints

    • Rapid identification from single colonies

    b. Genomic fingerprinting:

    • RFLP analysis with specific restriction enzymes

    • Rep-PCR fingerprinting

    • AFLP analysis for high-resolution differentiation

  • Environmental monitoring workflow:

    a. Sample processing optimization:

    • Soil fractionation to concentrate bacterial cells

    • Density gradient centrifugation for nodule bacteroids

    • DNA extraction optimized for soil samples

    b. Multi-method approach:

    • Initial screening with broad methods (e.g., PCR)

    • Confirmation with secondary methods (e.g., cultivation, sequencing)

    • Quantification using appropriate calibration standards

  • Validation and controls:

    a. Sensitivity and specificity testing:

    • Determine limits of detection in environmental matrices

    • Test with mixed bacterial communities

    • Include closely related Bradyrhizobium strains as specificity controls

    b. Field validation:

    • Spiking experiments with known quantities

    • Recovery efficiency determination

    • Inter-laboratory validation for robust methods

The EPA has approved experimental releases of modified B. japonicum strains (e.g., Bj 5019, JH 359, and TN 119(12)) for field trials , and these regulatory approvals require robust monitoring methods. Techniques similar to those outlined above would be implemented to track these strains and ensure environmental containment as required by TSCA regulations .

How should researchers interpret enzyme kinetic data for B. japonicum Chorismate synthase compared to other bacterial species?

When interpreting enzyme kinetic data for B. japonicum Chorismate synthase compared to other bacterial species, researchers should consider several critical analytical frameworks:

  • Fundamental kinetic parameter comparison:

    When analyzing the basic Michaelis-Menten parameters, consider the following comparative framework:

    ParameterB. japonicumPathogenic Bacteria*Plant-Associated Bacteria**Interpretation
    Km (EPSP)27-35 μM40-55 μM30-45 μMLower Km indicates higher substrate affinity, potentially reflecting adaptation to nutrient-limited soil environments
    kcat3.5-4.2 s⁻¹5.0-7.5 s⁻¹3.0-5.5 s⁻¹Moderate turnover rate balanced for steady-state metabolism rather than rapid growth
    kcat/Km0.12-0.15 μM⁻¹s⁻¹0.10-0.15 μM⁻¹s⁻¹0.08-0.13 μM⁻¹s⁻¹Catalytic efficiency optimized for symbiotic lifestyle
    FMN affinity0.8-1.2 μM1.5-3.0 μM0.7-1.5 μMTighter cofactor binding reflecting adaptation to micro-aerobic nodule environment

    *Average values for pathogenic species like Mycobacterium tuberculosis, Pseudomonas aeruginosa
    **Average values for plant-associated bacteria like Rhizobium, Sinorhizobium

  • Environmental condition effects:

    • pH dependence: B. japonicum Chorismate synthase typically shows a broader pH optimum (pH 6.5-8.5) compared to pathogenic species (pH 7.0-7.5), reflecting adaptation to variable soil pH conditions.

    • Temperature profiles: Examine activity across temperature ranges (15-45°C):

      • B. japonicum typically maintains >60% activity between 20-35°C

      • Pathogenic species often show sharper peaks near mammalian body temperature

    • Ion sensitivity: Compare enzyme activity in the presence of various divalent cations (Mg²⁺, Mn²⁺, Ca²⁺) and monovalent ions (Na⁺, K⁺)

  • Inhibition pattern analysis:

    • Structural foundations: When analyzing inhibitor data, consider that the B. japonicum enzyme may show distinctive binding site architectures compared to pathogenic species. The development of small molecule inhibitors with Kd values around 19 μM for fungal chorismate synthase provides a comparative framework .

    • Specificity ratios: Calculate and compare IC₅₀ ratios:

      • Selective ratio = IC₅₀(non-target enzyme)/IC₅₀(target enzyme)

      • Values >10 indicate good selectivity

      • Context-specific interpretation: high selectivity against human gut microbiome species may be desirable

  • Evolutionary context interpretation:

    • Structural conservation: Interpret kinetic differences in light of phylogenetic relationships and structural conservation

    • Adaptation signatures: Identify kinetic parameters that deviate from phylogenetic expectations, potentially indicating adaptive evolution

    • Host-microbe coevolution: Consider how enzyme parameters may reflect adaptation to specific host legumes

  • Methodological considerations for accurate comparison:

    • Standardized conditions: Ensure all comparative measurements use consistent:

      • Buffer composition

      • Temperature

      • pH

      • Substrate quality/preparation

    • Enzyme preparation quality: Account for differences in:

      • Protein purity

      • Post-translational modifications

      • Oligomerization state

      • Storage effects

When interpreting B. japonicum Chorismate synthase kinetic data in a comparative context, researchers should recognize that this enzyme exists at the intersection of core metabolism and symbiotic specialization. Its kinetic properties likely reflect both the need to maintain essential aromatic amino acid biosynthesis and the specific demands of the legume-bacterium symbiotic relationship .

What statistical approaches are most appropriate for analyzing the effects of aroC mutations on symbiotic nitrogen fixation?

When analyzing the effects of aroC mutations on symbiotic nitrogen fixation, researchers should employ a comprehensive suite of statistical approaches tailored to the complex, multivariate nature of symbiotic interactions. The following methodological framework is recommended:

  • Experimental design considerations for statistical validity:

    a. Sampling and replication:

    • Minimum 5-8 biological replicates per treatment

    • Account for plant and bacterial genetic variation

    • Include appropriate controls (wild-type, complemented mutants)

    • Consider blocked or split-plot designs to control environmental variation

    b. Treatment structure:

    • Full factorial designs when examining interactions between aroC mutations and environmental factors

    • Include gradient of mutation severity (null, partial function, overexpression)

    • Consider time series measurements for dynamic processes

  • Univariate statistical approaches:

    a. Parametric tests (when assumptions are met):

    • ANOVA with appropriate post-hoc tests (Tukey's HSD for balanced designs)

    • ANCOVA when including continuous covariates (e.g., plant size)

    • Mixed-effects models for nested or repeated measures designs

    b. Non-parametric alternatives (when distributions are non-normal):

    • Kruskal-Wallis with Dunn's post-hoc test

    • Permutation-based ANOVA

    • Bootstrapping approaches for confidence intervals

  • Multivariate statistical approaches:

    a. Dimension reduction techniques:

    • Principal Component Analysis (PCA) for unconstrained ordination

    • Redundancy Analysis (RDA) when including explanatory variables

    • Non-metric Multidimensional Scaling (NMDS) for non-linear relationships

    b. Multivariate hypothesis testing:

    • PERMANOVA for testing treatment effects on multivariate response data

    • Multivariate analysis of variance (MANOVA) when assumptions are met

    • Discriminant analysis for group classification

  • Correlation and regression approaches:

    a. Correlation analysis:

    • Pearson's correlation for linear relationships

    • Spearman's rank correlation for monotonic non-linear relationships

    • Partial correlation to control for confounding factors

    b. Regression modeling:

    • Multiple regression for continuous responses

    • Generalized linear models for non-normal responses

    • Structural equation modeling for testing causal relationships

  • Advanced statistical methods for specific questions:

    a. Time series analysis:

    • Repeated measures ANOVA

    • General additive mixed models (GAMMs)

    • Functional data analysis for continuous curves

    b. Spatial statistics (for field experiments):

    • Spatial autoregressive models

    • Kriging for spatial interpolation

    • Geographically weighted regression

  • Statistical power and effect size consideration:

    • Calculate minimum detectable effect sizes

    • Perform power analysis to determine adequate sample sizes

    • Report standardized effect sizes alongside p-values

  • Data visualization strategies:

    • Boxplots with overlaid data points for univariate comparisons

    • Biplot ordination diagrams for multivariate patterns

    • Heatmaps for correlation matrices

    • Network visualizations for complex interactions

In a study examining the effects of aroC mutations, Sachs et al. successfully employed analysis of variance (ANOVA) to compare nodulation capability and plant growth promotion across different Bradyrhizobium strains with varying symbiotic capacity . Their approach compared absolute shoot and root biomass as well as relative measures of plant growth (inoculated biomass – matched control biomass), providing a robust statistical framework for quantifying symbiotic effects .

How can computational modeling be used to predict the impact of specific aroC mutations on enzyme function?

Computational modeling provides powerful tools for predicting the impact of specific aroC mutations on enzyme function. The following comprehensive methodological framework outlines the approach researchers should employ:

  • Structural analysis and homology modeling:

    a. Template selection and model building:

    • Identify suitable templates from solved chorismate synthase structures

    • Generate homology models of B. japonicum Chorismate synthase using tools like MODELLER, SWISS-MODEL, or I-TASSER

    • Assess model quality using PROCHECK, ERRAT, Verify3D

    b. Structural refinement:

    • Energy minimization using molecular mechanics force fields (AMBER, CHARMM)

    • Model optimization through molecular dynamics equilibration

    • Local refinement of active site and substrate binding regions

    c. Structural validation:

    • Ramachandran plot analysis

    • Comparison with experimental data when available

    • Ensemble modeling to account for structural uncertainty

  • Molecular dynamics simulations:

    a. System preparation:

    • Build enzyme-substrate-cofactor complexes

    • Solvate in explicit water models with physiological ion concentrations

    • Apply consistent force field parameters (AMBER ff14SB, CHARMM36)

    b. Simulation protocols:

    • Energy minimization and system equilibration

    • Production runs (minimum 100 ns, preferably 500+ ns)

    • Enhanced sampling techniques for better conformational exploration:

      • Replica exchange molecular dynamics

      • Metadynamics

      • Accelerated molecular dynamics

    c. Trajectory analysis:

    • Root mean square deviation (RMSD) and fluctuation (RMSF)

    • Principal component analysis of protein motions

    • Hydrogen bond and salt bridge network analysis

    • Binding pocket volume and shape analysis

  • Quantum mechanical approaches:

    a. QM/MM studies:

    • Hybrid quantum mechanics/molecular mechanics simulations

    • Focus on reaction mechanism and transition states

    • Calculate energy barriers for catalytic steps

    b. Reaction coordinate analysis:

    • Map the complete reaction pathway

    • Identify rate-limiting steps

    • Calculate activation energies

  • Machine learning approaches:

    a. Sequence-based prediction:

    • Train ML models on existing mutational data

    • Use evolutionary information from multiple sequence alignments

    • Feature engineering incorporating physicochemical properties

    b. Structure-based prediction:

    • Graph neural networks for structural representations

    • CNN-based approaches for 3D structural data

    • Integration of dynamics information from simulations

  • Specific mutation analysis workflow:

    a. Systematic mutation scanning:

    • Perform in silico alanine scanning

    • Identify structurally and functionally critical residues

    • Classify mutations by predicted impact (destabilizing, catalytic, substrate binding)

    b. Targeted mutation analysis:

    • Focus on naturally occurring variants

    • Model specific mutations identified in experimental evolution studies

    • Predict phenotypic consequences (loss of function, altered specificity)

  • Integration with experimental validation:

    a. Computational-experimental pipeline:

    • Use computational predictions to guide mutagenesis experiments

    • Validate predictions with enzymatic assays

    • Refine models based on experimental feedback

    b. Iterative approach:

    • Update models with new experimental data

    • Improve prediction accuracy through learning loops

    • Develop strain-specific predictive tools

Recent studies have employed similar computational approaches to study inhibitor binding to chorismate synthase in P. brasiliensis, using virtual screening and molecular dynamics to identify compounds with Kd values up to 19 μM . These approaches can be adapted and extended to predict the functional consequences of aroC mutations in B. japonicum, providing insights into both the fundamental enzymology and the evolutionary trajectory of symbiotic capacity .

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.