Recombinant Bradyrhizobium japonicum 2,3,4,5-tetrahydropyridine-2,6-dicarboxylate N-succinyltransferase (dapD)

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 collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our default glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid 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, and we will prioritize its development.
Synonyms
dapD; blr8104; 2,3,4,5-tetrahydropyridine-2,6-dicarboxylate N-succinyltransferase; EC 2.3.1.117; Tetrahydrodipicolinate N-succinyltransferase; THDP succinyltransferase; THP succinyltransferase; Tetrahydropicolinate succinylase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-281
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bradyrhizobium diazoefficiens (strain JCM 10833 / IAM 13628 / NBRC 14792 / USDA 110)
Target Names
dapD
Target Protein Sequence
MSLSALESTI NSAFDARDGI STSTKGEVRE AVDQVLETLD KGEARVAERG ADGKWKVNQW LKKAVLLSFR LNDMGVIPGG PGQATWWDKV PSKFEGWGEN RFRDAGFRAV PGAVVRRSAF IAKNVVLMPS FVNLGAYVDE STMVDTWATV GSCAQIGKRV HISGGAGIGG VLEPLQAEPV IIEDDCFIGA RSEVAEGVIV RKGAVLAMGV FLGASTKIVD RDTGEVFIGE VPEYSVVVPG ALPGKPMKNG HIGPSTACAV IVKRVDERTR SKTSINELLR D
Uniprot No.

Target Background

Database Links

KEGG: bja:blr8104

STRING: 224911.blr8104

Protein Families
Transferase hexapeptide repeat family
Subcellular Location
Cytoplasm.

Q&A

What is the role of 2,3,4,5-tetrahydropyridine-2,6-dicarboxylate N-succinyltransferase (dapD) in Bradyrhizobium japonicum?

The dapD gene in Bradyrhizobium japonicum encodes for 2,3,4,5-tetrahydropyridine-2,6-dicarboxylate N-succinyltransferase, an enzyme that catalyzes the reaction of tetrahydrodipicolinate (THDP) and succinyl-CoA to form (S)-2-(3-carboxypropanamido)-6-oxoheptanedioic acid and coenzyme A. This enzyme plays a critical role in the diaminopimelate-lysine biosynthesis pathway, which produces two metabolites necessary for bacterial survival and growth: meso-diaminopimelate (m-DAP) for peptidoglycan synthesis and lysine for protein synthesis . The pathway is particularly important for B. japonicum's survival as a free-living soil bacterium and during symbiotic nitrogen fixation with soybean plants.

Why is dapD considered a potential target for antibiotic development?

DapD is considered a promising target for antibiotic development for several key reasons:

  • Essential role: DapD is crucial for bacterial cell wall synthesis and protein production through the diaminopimelate-lysine pathway.

  • Absence in humans: Since lysine is an essential amino acid for humans (we cannot synthesize it), this pathway doesn't exist in human cells, making dapD a potentially safe target for antibiotic therapies .

  • Conservation: The enzyme is conserved across many pathogenic bacterial species.

  • Structure: The mechanistic details and structural characteristics of dapD provide opportunities for rational drug design.

Research on B. japonicum dapD could provide insights applicable to developing antibiotics targeting this enzyme in pathogenic bacteria, while also improving our understanding of rhizobial metabolism .

What are the established methods for cloning the dapD gene from Bradyrhizobium japonicum?

When cloning the dapD gene from B. japonicum, researchers typically follow these methodological steps:

  • Genomic DNA extraction: Culture B. japonicum in HEPES-MES medium supplemented with 0.1% L-arabinose for 5 days at 28°C. Extract total DNA using BL extraction buffer following the protocol established by Hiraishi et al. .

  • PCR amplification: Design specific primers flanking the dapD gene based on the B. japonicum USDA 110 genome sequence. Use high-fidelity DNA polymerase (such as Ex Taq) with the following PCR cycling conditions:

    • Initial denaturation: 94°C for 5 min

    • 30 cycles of: 94°C for 30s, 55-60°C for 30s, 72°C for 1 min

    • Final extension: 72°C for 10 min

  • Cloning strategies: Use one of the following approaches:

    • Direct cloning into an expression vector (for immediate protein production)

    • Subcloning into an intermediate vector with subsequent transfer to an expression vector

    • Gateway cloning system for flexible construct generation

  • Vector selection: Common vectors for B. japonicum genes include pVK102, which has been successfully used for cloning rhizobial genes . For heterologous expression in E. coli, pET vectors are frequently employed.

  • Verification: Confirm cloned sequences by restriction analysis and DNA sequencing to ensure no mutations were introduced during amplification .

What are the optimal conditions for heterologous expression of B. japonicum dapD in E. coli?

For optimal heterologous expression of B. japonicum dapD in E. coli, consider the following protocol based on successful expression of related enzymes:

  • Host strain selection: BL21(DE3) or its derivatives are recommended for high-level expression. For complementation studies, E. coli dapD auxotrophs (such as strains with ΔdapD mutations) should be used .

  • Expression vector: Use T7 promoter-based vectors like pET series with appropriate affinity tags (His6 for ease of purification).

  • Growth conditions:

    • Medium: LB or 2xYT supplemented with appropriate antibiotics

    • Temperature: 28-30°C often yields better results than 37°C for B. japonicum proteins

    • Induction: 0.1-0.5 mM IPTG when culture reaches OD600 of 0.6-0.8

    • Post-induction: 16-18 hours at 16-18°C often improves protein solubility

  • Overcoming expression challenges:

    • Codon optimization may be necessary due to differing codon usage between B. japonicum and E. coli

    • Co-expression with chaperones (GroEL/GroES) can enhance proper folding

    • Fusion tags like MBP or SUMO may increase solubility of the recombinant protein

  • Functional validation: Verify enzymatic activity by complementation of E. coli dapD mutants. Growth restoration on minimal medium without diaminopimelic acid supplementation confirms functional expression .

What is the most effective purification strategy for recombinant B. japonicum dapD?

Based on successful purification protocols for similar enzymes, the following methodological approach is recommended:

  • Cell lysis: Harvest cells and resuspend in buffer containing 50 mM HEPES pH 7.5, 300 mM NaCl, 10% glycerol, 1 mM DTT, and protease inhibitors. Lyse cells using sonication or French press.

  • Initial purification: For His-tagged dapD, use Ni-NTA affinity chromatography:

    • Equilibrate column with binding buffer (50 mM HEPES pH 7.5, 300 mM NaCl, 10% glycerol)

    • Apply clarified lysate

    • Wash with binding buffer containing 20-30 mM imidazole

    • Elute with 250 mM imidazole

  • Secondary purification:

    • Ion exchange chromatography: Use Q-Sepharose column at pH 8.0

    • Size exclusion chromatography: Superdex 200 in 20 mM HEPES pH 7.5, 150 mM NaCl, 5% glycerol, 1 mM DTT

  • Quality assessment:

    • SDS-PAGE for purity (>95%)

    • Western blot for identity confirmation

    • Dynamic light scattering for homogeneity analysis

    • Mass spectrometry for accurate mass determination

  • Storage considerations: Store purified enzyme in 20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM DTT, 50% glycerol at -80°C for long-term stability .

How can I determine the kinetic parameters of recombinant B. japonicum dapD?

To determine the kinetic parameters of recombinant B. japonicum dapD, follow this methodological framework:

  • Assay selection:

    • Direct assay: Monitor the decrease in succinyl-CoA absorbance at 232 nm

    • Coupled assay: Link CoA formation to NAD+ reduction via appropriate coupling enzymes

  • Optimized reaction conditions:

    • Buffer: 50 mM HEPES pH 7.5

    • Temperature: 30°C (typical for B. japonicum enzymes)

    • Additives: 1-10 mM DTT, 5-10% glycerol

    • Metal ions: Test the effect of divalent cations (Mg²⁺, Ca²⁺) at 1-5 mM

  • Kinetic parameter determination:

    • Vary substrate concentrations (THDP and succinyl-CoA) independently

    • Plot initial velocities against substrate concentrations

    • Fit data to appropriate kinetic models (Michaelis-Menten, etc.)

    • Calculate KM, kcat, and kcat/KM values

  • Statistical analysis:

    • Perform experiments in triplicate

    • Calculate standard errors for all parameters

    • Use appropriate software (e.g., GraphPad Prism, Origin) for curve fitting

Based on studies of similar enzymes, expected parameters might be in the range of:

  • KM for THDP: 1-5 mM

  • KM for succinyl-CoA: 50-200 μM

  • kcat: 30-50 × 10⁻³ s⁻¹

How can I identify essential catalytic residues in B. japonicum dapD through site-directed mutagenesis?

To identify essential catalytic residues in B. japonicum dapD through site-directed mutagenesis, follow this systematic approach:

  • Target residue selection:

    • Compare B. japonicum dapD sequence with homologs from other bacteria

    • Identify highly conserved residues, particularly those in the active site

    • Focus on histidine, cysteine, aspartate, and glutamate residues that could participate in catalysis

  • Mutagenesis strategy:

    • Use site-directed mutagenesis techniques (such as QuikChange) to introduce specific mutations

    • Target conservative (e.g., Asp→Glu) and non-conservative (e.g., His→Ala) substitutions

    • Create a panel of mutants affecting different regions of the protein

  • Expression and purification:

    • Express wild-type and mutant proteins under identical conditions

    • Verify proper folding using circular dichroism spectroscopy

    • Confirm structural integrity through thermal denaturation experiments

  • Functional characterization:

    • Measure kinetic parameters (KM, kcat, kcat/KM) for each mutant

    • Compare with wild-type values to assess impact on catalysis

    • Determine if mutations affect substrate binding, catalytic efficiency, or both

  • In vivo validation:

    • Test the ability of mutant genes to complement E. coli dapD mutants

    • Quantify growth rates in minimal medium to assess in vivo enzyme functionality

This methodical approach will provide insights into the catalytic mechanism and structure-function relationships of B. japonicum dapD.

What is the most efficient approach for creating a dapD knockout mutant in Bradyrhizobium japonicum?

Creating a dapD knockout mutant in Bradyrhizobium japonicum requires careful planning due to the bacterium's slow growth and potential essentiality of the dapD gene. Here's a recommended methodological approach:

  • Construct design:

    • Create a suicide plasmid containing dapD gene fragments interrupted by an antibiotic resistance cassette (kanamycin or spectinomycin)

    • Ensure sufficient flanking homologous regions (>500 bp on each side) for efficient recombination

  • Transformation method:

    • Electroporation of B. japonicum competent cells with suicide plasmid

    • Alternatively, triparental mating using an E. coli donor strain carrying the construct, a helper strain with pRK2013, and B. japonicum recipient

  • Selection strategy:

    • Plate transformants on selective medium containing appropriate antibiotics

    • Due to the high incidence of spontaneous antibiotic resistance in B. japonicum, use a dual selection strategy with antibiotic markers and counter-selection

  • Mutant verification:

    • Colony PCR screening for initial identification

    • Southern blot hybridization to confirm proper integration

    • DNA sequencing to verify the exact nature of the mutation

  • Mutant viability considerations:

    • If dapD is essential, prepare conditional mutants using inducible promoters

    • Supplement growth medium with meso-diaminopimelic acid (m-DAP) and lysine

    • Characterize mutant phenotype in free-living conditions and during symbiosis with soybeans

A time-efficient method developed for B. japonicum involves:

  • Simple plate selection for antibiotic-resistant mutants

  • Colony streaking for isolation

  • DNA hybridization on nitrocellulose filters for direct identification of recombinant mutants
    This approach eliminates the need to first isolate genomic DNA from each potential mutant for Southern hybridization .

What factorial design approach should I use to optimize B. japonicum dapD enzyme assay conditions?

Optimizing B. japonicum dapD enzyme assay conditions can be achieved through a systematic factorial design approach following these methodological steps:

  • Select critical factors for initial screening:

    • Buffer composition (HEPES, TRIS, phosphate)

    • pH (range 6.5-8.5)

    • Temperature (25-40°C)

    • Divalent ions (Mg²⁺, Ca²⁺, concentrations 0-10 mM)

    • Salt concentration (NaCl, KCl, 0-500 mM)

    • Reducing agents (DTT, β-mercaptoethanol, 0-10 mM)

    • Solvent additives (DMSO, glycerol, 0-10%)

  • Implement a two-level factorial design:

    • Use Design-Expert software to generate a fractional factorial design

    • For eight factors, a 2⁸⁻⁴ design (16 runs) is recommended for initial screening

    • Include center points to detect curvature in the response surface

  • Analyze factor significance:

    • Generate half-normal probability plots to identify significant effects

    • Create Pareto charts to rank the importance of each factor

    • Identify interactions between variables affecting enzyme activity

  • Optimize significant factors:

    • Use response surface methodology (RSM) with central composite design (CCD)

    • Develop mathematical models describing the relationship between variables

    • Generate 3D response surface plots to visualize optimal conditions

  • Validate optimization results:

    • Perform confirmatory experiments under the predicted optimal conditions

    • Determine kinetic parameters (KM, Vmax, kcat) under optimized conditions

    • Calculate Z' factor to assess assay robustness (aim for Z' > 0.5)

An example dataset from a similar enzyme optimization study:

Table 1: Fractional Factorial Design for dapD Enzyme Assay Optimization

RunpHTemperature (°C)Reaction time (min)[MgCl₂] (mM)[NaCl] (mM)[DTT] (mM)[DMSO] (%)[Glycerol] (%)Activity (U/mL)
17.0253001001.002.0345
28.025305.01001.05.02.0412
37.037305.010010.002.0509
48.03730010010.05.02.0467
57.025605.030010.0010.0578
68.02560030010.05.010.0624
77.0376003001.05.010.0483
88.037605.03001.0010.0521
97.02530030010.05.010.0396
108.025305.030010.0010.0452
117.037305.03001.05.010.0487
128.0373003001.0010.0418
137.025605.01001.05.02.0563
148.0256001001.002.0498
157.03760010010.002.0625
168.037605.010010.05.02.0687

After analysis, optimal conditions might include: pH 7.5, 60 min reaction time, 9.2 mM DTT and 6.5% (v/v) glycerol, resulting in enzyme activity of approximately 730 U/mL .

How can I analyze the effects of copper ions on B. japonicum dapD activity and stability?

To systematically analyze the effects of copper ions on B. japonicum dapD activity and stability, follow this methodological framework:

  • Enzyme activity measurements:

    • Prepare enzyme in buffer without chelating agents

    • Incubate with varying Cu²⁺ concentrations (0-100 μM)

    • Measure residual activity using standard assay conditions

    • Calculate IC₅₀ values for enzyme inhibition

  • Kinetic analysis of inhibition:

    • Determine enzyme activity at different substrate concentrations with varying Cu²⁺ levels

    • Analyze data using Lineweaver-Burk plots to identify inhibition type (competitive, non-competitive, uncompetitive)

    • Calculate inhibition constants (Ki)

  • Protection studies:

    • Test protective effects of reducing agents (DTT, 1-10 mM)

    • Examine protection by chelating agents (EDTA, 1-10 mM)

    • Assess protection by substrate binding

  • Structural analysis:

    • Monitor tryptophan fluorescence quenching by Cu²⁺

    • Calculate binding affinity (KD) from fluorescence data

    • Compare spectra with chemically denatured enzyme to assess structural changes

    • Use circular dichroism to observe secondary structure alterations

  • Metal binding characterization:

    • Employ EPR spectroscopy to identify Cu²⁺ binding sites

    • Determine if Cu²⁺ is reduced to Cu⁺ during interaction

    • Identify potential binding residues (histidine, cysteine)

Expected results based on similar enzymes suggest that Cu²⁺ may rapidly and specifically inactivate dapD at low concentrations (KD ≈ 2-3 μM), with activity protected in reducing environments. Tryptophan fluorescence quenching and EPR data may indicate binding to histidine residues and potential reduction to Cu⁺, suggesting an oxidative inactivation mechanism that results in significant structural changes .

How can I design B. japonicum dapD mutants with enhanced catalytic efficiency through directed evolution?

Designing B. japonicum dapD mutants with enhanced catalytic efficiency through directed evolution requires a systematic approach:

  • Library generation strategies:

    • Error-prone PCR: Use unbalanced dNTP concentrations and MnCl₂ to introduce random mutations

    • DNA shuffling: Fragment and reassemble dapD genes from different bacterial species

    • Site-saturation mutagenesis: Target active site residues with NNK codons

    • Create a mutator strain: Introduce the dapD gene into B. japonicum with a mutated dnaQ gene (encoding DNA polymerase III epsilon subunit) to increase mutation rate

  • Selection system design:

    • Develop a selection strategy using E. coli ΔdapD auxotrophs

    • Create selection medium containing gradually decreasing concentrations of meso-diaminopimelic acid

    • Include competitive growth conditions to select for most efficient variants

  • Screening methodology:

    • Develop a high-throughput colorimetric assay for dapD activity

    • Use 96-well plate format to screen hundreds to thousands of variants

    • Implement multi-tier screening approach:

      • Tier 1: Growth-based selection for functional variants

      • Tier 2: Activity screening for improved catalytic efficiency

      • Tier 3: Detailed kinetic characterization of promising candidates

  • Iterative optimization:

    • Perform multiple rounds of mutation and selection

    • Recombine beneficial mutations from different variants

    • Use machine learning to predict promising mutation combinations

  • Structural and functional validation:

    • Determine crystal structures of improved variants

    • Compare active site architecture with wild-type enzyme

    • Analyze structural basis for enhanced catalysis

Successful directed evolution could potentially achieve:

  • 10-100 fold improvement in kcat/KM values

  • Enhanced stability under industrial conditions

  • Altered substrate specificity for biotechnological applications

What approaches can I use to study the role of B. japonicum dapD in symbiotic nitrogen fixation with soybeans?

To comprehensively study the role of B. japonicum dapD in symbiotic nitrogen fixation with soybeans, implement this multi-faceted methodological approach:

  • Genetic manipulation strategies:

    • Create conditional dapD mutants using inducible promoters

    • Develop strains with varying dapD expression levels

    • Complement mutants with wild-type and modified dapD genes

  • Symbiosis establishment analysis:

    • Inoculate soybean plants with wild-type and mutant strains

    • Quantify nodulation kinetics, nodule number, and nodule morphology

    • Assess competitiveness through co-inoculation experiments with wild-type strains

    • Use a defined mixing ratio (e.g., 1:1) and identify bacteria in nodules using strain-specific markers

  • Bacteroid development assessment:

    • Isolate bacteroids from nodules at different developmental stages

    • Measure dapD expression using RT-qPCR and Western blot analysis

    • Determine peptidoglycan composition and cell wall integrity

    • Analyze bacteroid morphology using electron microscopy

  • Nitrogen fixation measurement:

    • Quantify nitrogenase activity using acetylene reduction assay

    • Measure leghemoglobin content in nodules

    • Assess plant growth parameters (shoot dry weight, nitrogen content)

    • Compare wild-type with dapD mutant and complemented strains

  • Metabolic profiling:

    • Conduct comparative metabolomics of free-living bacteria and bacteroids

    • Focus on lysine, diaminopimelate, and related metabolites

    • Analyze carbon flux through central metabolic pathways

    • Determine impacts on other symbiosis-related pathways

  • Transcriptomic analysis:

    • Perform RNA-seq to examine global gene expression changes

    • Compare expression profiles between wild-type and mutant strains

    • Identify regulatory networks involving dapD

    • Examine coordination with other symbiosis genes (nif, fix, nod)

Expected outcomes may reveal that dapD is crucial for bacteroid development and maintenance, potentially affecting both peptidoglycan remodeling during bacteroid differentiation and lysine biosynthesis for protein synthesis during symbiotic nitrogen fixation .

What statistical approaches should I use to analyze dapD enzyme kinetics data with potential outliers?

When analyzing dapD enzyme kinetics data with potential outliers, employ this comprehensive statistical framework:

  • Data preprocessing and visualization:

    • Plot raw data using scatter plots and residual plots to identify potential outliers

    • Create Michaelis-Menten, Lineweaver-Burk, and Eadie-Hofstee plots to visualize kinetic behavior

    • Use box plots to identify values outside the interquartile range

  • Outlier identification methods:

    • Apply Grubbs' test for single outliers (α = 0.05)

    • Use Dixon's Q test for small sample sets

    • Implement Chauvenet's criterion for larger datasets

    • Apply ROUT method (Robust regression and Outlier removal) with Q = 1%

  • Robust regression approaches:

    • Use weighted non-linear regression with reduced weight for outlier points

    • Apply iteratively reweighted least squares method

    • Employ bisquare or Huber weighting functions to minimize outlier influence

    • Utilize GraphPad Prism or R software packages for implementation

  • Model selection criteria:

    • Calculate Akaike Information Criterion (AIC) and Bayesian Information Criterion (BIC)

    • Compare competitive models (different kinetic models with/without outliers)

    • Use F-test to determine if complex models provide statistically better fits

  • Validation and reporting:

    • Report both analyses with and without outliers for transparency

    • Include 95% confidence intervals for all kinetic parameters

    • Validate findings with bootstrap resampling (1000+ iterations)

    • Present residual plots to demonstrate goodness of fit

For a comprehensive data analysis plan, follow the structure outlined in the VA HSR&D Data Analysis Plan instructions, including detailed methods for handling missing data, definitions of analytical sets, and plans for covariates and data transformations .

How should I design experiments to resolve contradictory results between in vitro and in vivo studies of B. japonicum dapD function?

When confronted with contradictory results between in vitro and in vivo studies of B. japonicum dapD function, implement this systematic experimental design approach:

  • Critical evaluation of methodological differences:

    • Create a detailed comparison table of experimental conditions

    • Identify key variables that differ between in vitro and in vivo systems

    • Assess potential confounding factors in both experimental setups

    • Formulate testable hypotheses to explain discrepancies

  • Bridging experiments design:

    • Develop intermediate experimental systems to bridge the gap:

      • Cell-free extracts from B. japonicum cultures

      • Permeabilized cells retaining cellular organization

      • Ex planta nodule extracts maintaining symbiotic conditions

    • Match buffer conditions, pH, and ionic strength to physiological environments

  • Controlled variable manipulation:

    • Implement a discrete choice experiment (DCE) approach:

      • Define key attributes that may affect enzyme behavior

      • Create experimental designs testing combinations of these attributes

      • Use factorial design to identify interaction effects

      • Analyze using multinomial logit models

  • Multi-method validation:

    • Apply orthogonal experimental techniques to measure the same parameters

    • For kinetic studies, use different detection methods (spectrophotometric, HPLC, coupled assays)

    • For in vivo studies, combine genetic, biochemical, and physiological approaches

  • Advanced analytical approaches:

    • Implement Bayesian statistical analysis to incorporate prior knowledge

    • Use D-efficient experimental designs for optimal parameter estimation

    • Apply D₂-efficient designs when resolving complex interaction effects

  • Systematic reporting framework:

    • Document all experimental variables according to STRENDA guidelines

    • Report data from intermediate experiments that bridge the gap

    • Address limitations transparently

    • Present unified models that explain both sets of results

Table 2: Framework for Resolving In Vitro vs. In Vivo Discrepancies

ParameterIn Vitro ConditionIn Vivo ConditionBridging ApproachExpected Outcome
pHControlled (7.5)Variable (6.5-7.5)pH titration experimentsIdentify pH optimum and physiological range
Ionic strengthDefined buffersComplex cytoplasmic environmentGradual addition of cellular extractsDetermine effect of cellular milieu
Substrate availabilitySaturatingLimitingConcentration series at physiological rangesEstablish realistic kinetic parameters
Post-translational modificationsAbsentPresentAnalysis of purified protein from bacteroidsIdentify regulatory modifications
Protein-protein interactionsIsolated enzymeInteraction networksPull-down assays and interactome analysisMap functional interaction partners

This approach systematically narrows the gap between controlled in vitro conditions and complex in vivo environments, providing a more complete understanding of B. japonicum dapD function in its native context .

How can emerging deep learning approaches be applied to improve B. japonicum dapD structure prediction and function analysis?

Emerging deep learning approaches offer powerful tools for improving B. japonicum dapD structure prediction and functional analysis:

  • Advanced structure prediction methods:

    • Implement AlphaFold2 or RoseTTAFold to generate high-accuracy structural models

    • Use multiple sequence alignments of DapD homologs to enhance prediction accuracy

    • Apply specific refinement protocols for active site regions

    • Validate predictions through comparison with homologous crystal structures

  • Molecular dynamics simulations:

    • Conduct long-timescale MD simulations (>100 ns) using GPU acceleration

    • Apply machine learning-based force fields for improved accuracy

    • Analyze conformational dynamics of substrate binding and catalytic events

    • Identify allosteric sites and communication pathways through network analysis

  • Deep learning for functional site prediction:

    • Train neural networks on known enzyme active sites to predict catalytic residues

    • Use graph convolutional networks to analyze protein structure as a spatial graph

    • Apply attention mechanisms to focus on evolutionarily conserved regions

    • Validate predictions through site-directed mutagenesis experiments

  • Protein-ligand interaction modeling:

    • Employ deep learning-based scoring functions for molecular docking

    • Utilize generative adversarial networks (GANs) to design novel inhibitors

    • Predict binding affinities using graph neural networks

    • Validate in silico predictions with experimental binding assays

  • Integration with experimental data:

    • Use Bayesian frameworks to incorporate experimental constraints

    • Apply transfer learning from related enzymes with known structures

    • Implement active learning approaches to guide experimental design

    • Develop hybrid models combining computational predictions with sparse experimental data

  • Interpretable AI for mechanism elucidation:

    • Apply explainable AI techniques to identify key structural determinants of function

    • Develop attention visualization methods to highlight critical residues

    • Use deep learning to classify enzyme variants based on kinetic parameters

    • Create mechanistic models incorporating quantum mechanical calculations for transition states

These advanced computational approaches can significantly accelerate understanding of B. japonicum dapD structure-function relationships and enable rational design of improved enzyme variants for biotechnological applications .

What new methodologies are being developed to study the impact of B. japonicum dapD on plant-microbe interactions in agricultural settings?

Cutting-edge methodologies for studying B. japonicum dapD's impact on plant-microbe interactions in agricultural settings include:

  • Advanced imaging technologies:

    • Apply light sheet microscopy for 3D visualization of nodule development

    • Use confocal Raman microscopy to map metabolite distributions in planta

    • Implement correlative light and electron microscopy (CLEM) to link function with ultrastructure

    • Apply expansion microscopy for super-resolution imaging of bacteroid cell walls

  • Multi-omics integration approaches:

    • Combine transcriptomics, proteomics, and metabolomics data from both partners

    • Implement network analysis to identify key regulatory hubs

    • Apply machine learning for pattern recognition across diverse datasets

    • Use stable isotope labeling to track metabolic fluxes between plant and bacteria

  • Field-deployable phenotyping technologies:

    • Develop portable devices for real-time monitoring of nitrogenase activity

    • Apply hyperspectral imaging to assess plant nitrogen status

    • Use IoT-based sensors for continuous field monitoring

    • Implement drone-based imaging for high-throughput phenotyping

  • CRISPR-based methodologies:

    • Develop CRISPRi systems for conditional knockdown of dapD in the field

    • Apply CRISPR-Cas base editors for precise mutagenesis without double-strand breaks

    • Use CRISPR screening to identify genetic interactions with dapD

    • Implement CRISPR activation systems to upregulate dapD expression

  • Synthetic biology approaches:

    • Engineer synthetic regulatory circuits controlling dapD expression

    • Develop optogenetic tools for spatiotemporal control of protein function

    • Create biosensors for real-time monitoring of lysine/DAP levels

    • Apply minimal genome approaches to isolate essential dapD functions

  • Advanced microbiome analysis:

    • Use microfluidic devices for single-cell isolation and analysis

    • Apply spatial transcriptomics to map gene expression in nodule sections

    • Implement meta-proteomics to study protein expression in complex communities

    • Develop synthetic communities with defined strains to dissect ecological interactions

  • Academic-industry collaborative frameworks:

    • Establish data sharing platforms for large-scale field trials

    • Develop standardized protocols for assessing symbiotic efficiency

    • Create repositories of characterized strain variants for agricultural applications

    • Implement FAIR (Findable, Accessible, Interoperable, Reusable) data principles

These emerging methodologies promise to transform our understanding of B. japonicum dapD's role in symbiotic nitrogen fixation and enable development of improved inoculants for sustainable agriculture .

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.