Recombinant Nitrosomonas europaea Shikimate dehydrogenase, also known as aroE, is an enzyme produced through recombinant DNA technology. This enzyme is crucial in the shikimate pathway, which is essential for the biosynthesis of aromatic compounds in microorganisms and plants. The shikimate pathway is absent in metazoans, making it a target for developing nontoxic herbicides and drugs .
Shikimate dehydrogenase catalyzes the fourth step of the shikimate pathway, converting 3-dehydroshikimate to shikimate. This step is vital for the synthesis of aromatic amino acids and other aromatic compounds necessary for microbial growth and metabolism .
Recombinant Nitrosomonas europaea Shikimate dehydrogenase (aroE) is produced in various host organisms such as yeast, E. coli, and mammalian cells . The choice of host can affect the enzyme's yield, purity, and stability. For instance, yeast is commonly used for its ability to perform complex post-translational modifications, while E. coli is favored for its high yield and ease of production .
The biochemical properties of recombinant Nitrosomonas europaea Shikimate dehydrogenase (aroE) include its specificity for NADP+ as a cofactor, similar to other shikimate dehydrogenases. The enzyme's activity is crucial for the conversion of 3-dehydroshikimate to shikimate, utilizing NADPH as a reducing agent .
Recombinant Nitrosomonas europaea Shikimate dehydrogenase (aroE) has potential applications in biotechnology and pharmaceutical research. It can be used to study the shikimate pathway in detail, which is important for understanding microbial metabolism and developing new antimicrobial agents .
Recombinant Nitrosomonas europaea Shikimate dehydrogenase (aroE) is a valuable tool for studying microbial metabolism and developing new biotechnological applications. Its production in various hosts allows for flexibility in research and potential industrial applications. Further research into this enzyme could provide insights into the shikimate pathway and its role in microbial biology.
KEGG: neu:NE1627
STRING: 228410.NE1627
Shikimate dehydrogenase (SDH) catalyzes the fourth step of the shikimate pathway, specifically the reversible NADPH-dependent reduction of 3-dehydroshikimate to shikimate. This reaction is essential for the biosynthesis of aromatic amino acids in plants, fungi, bacteria, and apicomplexan parasites . In Nitrosomonas europaea, an obligate chemolithoautotroph that derives energy from ammonia oxidation, the aroE gene encodes this critical enzyme that contributes to cellular biosynthetic processes despite the organism's limited capacity for organic compound catabolism . The shikimate pathway represents one of the few anabolic routes required for aromatic amino acid synthesis in this specialized bacterium.
While no crystal structure specific to N. europaea aroE is directly mentioned in the search results, comparative analysis with other SDH family members suggests significant structural conservation. Shikimate dehydrogenases typically display a similar architecture with two α/β domains separated by a wide cleft . The four main SDH classes (AroE, YdiB, SDH-like, and AroE-like1) share a high level of structural conservation extending to their active sites . Based on this conservation pattern, we can reasonably infer that N. europaea aroE likely exhibits the characteristic two-domain structure with a substrate-binding cleft and conformational flexibility that allows for movement between open and closed states upon substrate binding .
For expressing recombinant N. europaea aroE, Escherichia coli-based expression systems are typically most effective due to their high yield and ease of genetic manipulation. Based on studies with other shikimate dehydrogenases, the pET expression system under the control of the T7 promoter often provides optimal expression levels . When designing the expression construct, researchers should consider:
Codon optimization for E. coli, as N. europaea has a GC content that differs from E. coli
Addition of affinity tags (His6 is common) to facilitate purification
Inclusion of cleavage sites for tag removal if needed for structural studies
Expression at lower temperatures (16-20°C) to enhance protein solubility
E. coli BL21(DE3) or its derivatives are recommended host strains due to their reduced protease activity and compatibility with T7 expression systems. Induction conditions should be optimized with varying IPTG concentrations (0.1-1.0 mM) and induction times (4-16 hours) to maximize soluble protein yield.
A multi-step purification approach is recommended to achieve high purity recombinant N. europaea aroE:
| Purification Step | Method | Buffer System | Expected Result |
|---|---|---|---|
| Initial Capture | Immobilized Metal Affinity Chromatography (IMAC) | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10-250 mM imidazole gradient | 80-90% purity |
| Intermediate Purification | Ion Exchange Chromatography | 20 mM Tris-HCl pH 8.0, 0-500 mM NaCl gradient | 90-95% purity |
| Polishing | Size Exclusion Chromatography | 20 mM Tris-HCl pH 8.0, 150 mM NaCl | >98% purity |
Throughout purification, adding reducing agents (1-5 mM DTT or 2-10 mM β-mercaptoethanol) and protease inhibitors is advisable to maintain enzyme stability and activity. Samples from each purification step should be analyzed using SDS-PAGE to assess purity and Western blotting to confirm identity. Activity assays monitoring NADPH oxidation spectrophotometrically at 340 nm should be performed to track specific activity through the purification process.
Comprehensive stability and activity assessment of purified recombinant N. europaea aroE should include:
Thermal stability analysis:
Differential scanning fluorimetry (DSF) to determine melting temperature (Tm)
Activity retention after incubation at various temperatures (4-50°C)
Storage stability testing:
Activity monitoring during storage at different temperatures (-80°C, -20°C, 4°C)
Effects of cryoprotectants (glycerol 10-50%, sucrose, trehalose)
Freeze-thaw stability over multiple cycles
Activity characterization:
Determine optimal pH range (pH 5.0-9.0)
Establish kinetic parameters (Km, kcat, kcat/Km) for both shikimate and NADP+
Assess cofactor specificity (NADP+ vs. NAD+)
Determine substrate specificity using shikimate analogues
Structural integrity verification:
Circular dichroism (CD) spectroscopy to assess secondary structure
Dynamic light scattering (DLS) to verify monodispersity
Size exclusion chromatography with multi-angle light scattering (SEC-MALS) to determine oligomeric state
These analyses will establish baseline characteristics of the recombinant enzyme and ensure its suitability for downstream applications in structural or inhibitor studies.
For structural determination of N. europaea aroE, X-ray crystallography remains the gold standard approach, as demonstrated by successful structure determinations of other SDH family members at resolutions ranging from 1.5 to 2.5 Å . A systematic approach should include:
Crystallization screening:
Utilize commercial sparse matrix screens with varying precipitants, buffers, and additives
Test protein concentrations between 5-15 mg/ml
Screen with and without cofactors (NADP+/NADPH) and substrate/product
Implement sitting-drop and hanging-drop vapor diffusion methods
Crystal optimization:
Fine-tune promising conditions by varying pH (±0.5 units), precipitant concentration (±5%), and temperature
Add small molecule additives that may enhance crystal quality
Implement seeding techniques for improved nucleation control
Data collection and processing:
Collect high-resolution diffraction data at synchrotron radiation sources
Process data using standard crystallographic software packages
Solve structure by molecular replacement using other SDH family members as templates
Refine the structure to achieve optimal R-factors and geometry
Complementary approaches:
Cryo-electron microscopy (cryo-EM) for larger complexes or challenging crystallization cases
Nuclear magnetic resonance (NMR) for studying dynamic regions and ligand interactions
Small-angle X-ray scattering (SAXS) for solution structure and conformational changes
These approaches should be complemented with computational modeling to predict regions of conformational flexibility that might be observed between open and closed conformations during substrate binding, as noted in other SDH family members .
The catalytic mechanism of N. europaea aroE likely follows the conserved mechanism observed across the SDH family. Key components of this mechanism include:
Catalytic dyad: The active site contains a conserved lysine-aspartate pair that functions as a catalytic dyad rather than just as binding residues . This dyad is critical for proton abstraction during catalysis.
Substrate recognition: Based on sequence analysis and structural comparison across SDH family members, specific residues are involved in 3-dehydroshikimate recognition and binding .
Conformational changes: There is evidence suggesting that SDH enzymes undergo a conformational switch between open and closed states upon substrate binding . This conformational flexibility is likely conserved in N. europaea aroE.
Cofactor specificity: Different SDH family members exhibit varying cofactor preferences. While AroE is typically NADP-specific, YdiB can utilize either NAD or NADP . The specificity in N. europaea aroE would depend on specific residues in its dinucleotide-binding domain.
The conservation of the catalytic mechanism across phylogenetically distant SDH family members provides a strong foundation for predicting the mechanism in N. europaea aroE, though specific kinetic studies would be necessary to confirm these predictions and identify any unique features.
Based on comparative analysis with other SDH family members, several functional residues are likely critical in N. europaea aroE:
Site-directed mutagenesis studies targeting these residues would be valuable for confirming their roles in N. europaea aroE. Particularly, mutations of the catalytic lysine and aspartate residues would be expected to dramatically reduce enzymatic activity, while mutations in substrate binding residues might alter substrate specificity or affinity without completely abolishing activity.
The shikimate pathway represents an attractive target for antimicrobial development because it is present in bacteria, fungi, and apicomplexan parasites but absent in metazoans (animals) . This makes it an ideal target for developing compounds with selective toxicity. Recombinant N. europaea aroE can be utilized in several approaches for antimicrobial development:
High-throughput screening (HTS):
Develop a robust assay monitoring NADPH oxidation spectrophotometrically
Screen diverse chemical libraries against purified recombinant enzyme
Establish counter-screens to eliminate compounds affecting assay components
Structure-based drug design:
Use the crystal structure (once determined) for virtual screening
Implement molecular docking to identify compounds that bind the active site
Design transition-state analogues based on the catalytic mechanism
Fragment-based approach:
Screen fragment libraries using thermal shift assays or NMR
Elaborate hit fragments guided by structural information
Link fragments that bind to different pockets
Comparative analysis with other pathogens:
Explore the conservation of binding sites across pathogenic species
Identify selective inhibitors that target specific pathogen enzymes
Develop broad-spectrum inhibitors targeting highly conserved features
The conservation of three-dimensional fold, active site architecture, and catalytic mechanism among members of the SDH family will facilitate the design of drugs targeting multiple pathogens through the shikimate pathway .
Systematic mutagenesis approaches can reveal critical aspects of N. europaea aroE function and evolution:
Alanine scanning mutagenesis:
Systematically replace conserved residues with alanine
Measure kinetic parameters of mutants to identify critical residues
Map functional hotspots on the structure
Ancestral sequence reconstruction:
Infer ancestral sequences of aroE across bacterial lineages
Express and characterize reconstructed ancestral enzymes
Identify key mutations that led to functional divergence
Domain swapping experiments:
Create chimeric enzymes by swapping domains between N. europaea aroE and other SDH family members
Determine which domains control substrate specificity, cofactor preference, and catalytic efficiency
Understand the modular nature of enzyme evolution
Directed evolution:
Create libraries with random or site-directed mutations
Select for variants with enhanced activity, stability, or altered specificity
Sequence beneficial variants to identify unexpected functional residues
These approaches can provide insights into how the four functionally distinct enzyme classes in the SDH family (AroE, YdiB, SDH-like, and AroE-like1) evolved and diverged , and what unique adaptations N. europaea might have developed in its aroE enzyme.
Understanding aroE function within the metabolic context of N. europaea requires consideration of this organism's unique chemolithoautotrophic lifestyle:
N. europaea has a streamlined genome with limited genes for catabolism of organic compounds but maintains essential biosynthetic pathways . Understanding how aroE functions within this specialized metabolic network provides insights into the adaptations of obligate chemolithoautotrophs to their ecological niche.
Solubility and stability challenges are common when working with recombinant enzymes. For N. europaea aroE, consider the following strategies:
Enhancing solubility during expression:
Lower induction temperature (16-20°C)
Reduce inducer concentration
Co-express with molecular chaperones (GroEL/GroES, DnaK/DnaJ/GrpE)
Use solubility-enhancing fusion partners (MBP, SUMO, TrxA)
Optimize codon usage for expression host
Buffer optimization for stability:
Screen different buffer systems (HEPES, Tris, phosphate) at pH 6.5-8.5
Test stabilizing additives:
Osmolytes (glycerol, sorbitol, trehalose)
Salts (NaCl, KCl, (NH4)2SO4 at 50-500 mM)
Reducing agents (DTT, β-mercaptoethanol, TCEP)
Divalent cations (Mg2+, Mn2+)
Include cofactors (NADP+) at low concentrations
Storage and handling:
Determine optimal protein concentration (avoid too dilute or concentrated)
Evaluate freeze-thaw stability and develop aliquoting strategies
Test lyophilization with appropriate excipients
Consider immobilization techniques for enhanced stability
Structural modifications:
Identify and mutate surface-exposed hydrophobic residues
Introduce disulfide bonds to stabilize tertiary structure
Remove flexible loops that may contribute to aggregation
Create truncated constructs if terminal regions cause instability
Implementation of these strategies should be systematic, testing one variable at a time and assessing effects on solubility, stability, and activity to identify optimal conditions for working with recombinant N. europaea aroE.
When encountering discrepancies in kinetic data for N. europaea aroE, consider these systematic troubleshooting approaches:
Assay validation and standardization:
Verify assay linearity with respect to time and enzyme concentration
Establish standard operating procedures for consistent measurements
Implement internal controls and reference standards
Ensure all reagents are fresh and of consistent quality
Enzyme quality assessment:
Confirm protein purity by orthogonal methods (SDS-PAGE, SEC, DLS)
Determine the proportion of active enzyme using active site titration
Assess oligomeric state and potential aggregation
Verify absence of co-purifying contaminants that might affect activity
Environmental variable control:
Strictly control temperature during measurements
Buffer all solutions to consistent pH
Eliminate oxidative damage by including reducing agents
Account for potential metal ion effects
Data analysis refinement:
Apply appropriate kinetic models (Michaelis-Menten, allosteric, bi-bi)
Use global fitting approaches for multi-parameter determination
Account for substrate/product inhibition
Implement statistical analysis to identify outliers
Advanced analytical approaches:
Employ isothermal titration calorimetry (ITC) for direct binding measurements
Use stopped-flow techniques for measuring rapid kinetics
Implement NMR for detecting structural changes during catalysis
Develop mass spectrometry approaches for monitoring reaction progress
By systematically addressing these aspects, researchers can identify sources of variability in kinetic measurements and develop robust protocols that yield consistent, reproducible data.
Investigating enzyme-enzyme interactions within the shikimate pathway requires a multi-faceted approach:
Co-immunoprecipitation and pull-down assays:
Express tagged versions of aroE and other pathway enzymes
Perform pull-down experiments to identify interacting partners
Verify interactions by reverse pull-down with different tags
Use crosslinking to stabilize transient interactions
Biophysical interaction analysis:
Surface plasmon resonance (SPR) to measure binding kinetics
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Microscale thermophoresis (MST) for solution-based interaction studies
Analytical ultracentrifugation to detect complex formation
Structural approaches:
Crystallize enzyme complexes to obtain structural information
Use small-angle X-ray scattering (SAXS) for solution structure of complexes
Implement crosslinking mass spectrometry (XL-MS) to map interaction interfaces
Apply cryo-electron microscopy for larger complexes
Functional studies:
Measure kinetic parameters in the presence of other pathway enzymes
Investigate substrate channeling between consecutive enzymes
Assess allosteric regulation by metabolites or pathway components
Study the effects of site-directed mutations at potential interaction interfaces
In vivo approaches:
Implement fluorescence resonance energy transfer (FRET) to detect interactions
Use bacterial two-hybrid systems for protein interaction screening
Perform co-localization studies using fluorescent tags
Apply proximity-dependent labeling methods (BioID, APEX)
Understanding these interactions is crucial as they may reveal metabolic channeling mechanisms and regulatory networks that coordinate flux through the shikimate pathway in N. europaea, potentially uncovering unique adaptations in this chemolithoautotroph compared to heterotrophic bacteria.