Recombinant Nitrosomonas europaea 4-diphosphocytidyl-2-C-methyl-D-erythritol kinase (ispE) is a recombinant protein derived from the bacterium Nitrosomonas europaea. This enzyme plays a crucial role in the biosynthesis pathway of isoprenoids, which are essential for various cellular functions. The ispE enzyme specifically catalyzes the phosphorylation of the position 2 hydroxy group of 4-diphosphocytidyl-2-C-methyl-D-erythritol, a key step in the production of isoprenoids via the non-mevalonate pathway .
The recombinant ispE enzyme is produced in different host organisms, including Escherichia coli and yeast. The enzyme from E. coli has a purity of more than 85% as determined by SDS-PAGE, and its sequence is well-documented . The enzyme's stability and shelf life depend on storage conditions, with a typical shelf life of 6 months for liquid forms stored at -20°C or -80°C and up to 12 months for lyophilized forms under the same conditions .
| Production Host | Purity | Shelf Life |
|---|---|---|
| E. coli | >85% | 6 months (liquid), 12 months (lyophilized) at -20°C/-80°C |
| Yeast | High purity | Not specified |
The ispE enzyme is involved in the non-mevalonate pathway, which is crucial for the synthesis of isoprenoids. Isoprenoids are vital for various cellular processes, including the synthesis of membrane components and signaling molecules. In Nitrosomonas europaea, this pathway supports the bacterium's growth and survival by providing essential lipids and other isoprenoid-derived compounds.
The recombinant ispE enzyme could have applications in biotechnology, particularly in the production of isoprenoid-derived compounds. These compounds have diverse applications, ranging from pharmaceuticals to biofuels. Further research is needed to explore the potential uses of this enzyme in industrial processes.
KEGG: neu:NE1827
STRING: 228410.NE1827
IspE (4-diphosphocytidyl-2-C-methyl-D-erythritol kinase) catalyzes the ATP-dependent phosphorylation of the 2-hydroxyl group of 4-diphosphocytidyl-2C-methyl-D-erythritol, forming 4-diphosphocytidyl-2C-methyl-D-erythritol 2-phosphate . This reaction represents the fourth step in the MEP pathway for isoprenoid biosynthesis, which is essential for the production of various terpenoids and components of cell membranes.
The catalytic mechanism involves:
Binding of ATP and the 4-diphosphocytidyl-2C-methyl-D-erythritol substrate
Nucleophilic attack of the 2-hydroxyl group on the γ-phosphate of ATP
Transfer of the phosphate group
Release of the phosphorylated product and ADP
The enzyme belongs to the GHMP kinase family (named after galactokinase, homoserine kinase, mevalonate kinase, and phosphomevalonate kinase), specifically the IspE subfamily .
Nitrosomonas europaea is a well-characterized ammonia-oxidizing bacterium and serves as a model organism for studying nitrification processes . Studying ispE from this organism offers several advantages:
N. europaea has a fully sequenced genome, facilitating genetic manipulation and protein expression studies
As an ammonia oxidizer that thrives in both oxygen-replete and oxygen-limited environments, N. europaea provides insights into enzyme adaptations under different metabolic conditions
The organism's ecological importance in nitrogen cycling makes its metabolic pathways, including isoprenoid biosynthesis, relevant to environmental microbiology
Comparison of N. europaea ispE with homologs from other organisms can reveal evolutionary adaptations in enzyme function
N. europaea ispE consists of 278 amino acids with a partial sequence beginning with MDIFPAPAKLNLFLHVIGRREDGYHLLQTVFRFIDHSDRLHFDITHDGVIRHENLIPGLTETDDLCVRAAKLL . Though the complete crystal structure of N. europaea ispE has not been reported in the provided search results, based on other characterized ispE enzymes, it likely features:
A typical GHMP kinase fold with distinct N-terminal and C-terminal domains
A central ATP-binding site with conserved motifs for phosphate binding
A substrate-binding pocket that accommodates 4-diphosphocytidyl-2C-methyl-D-erythritol
Critical residues for catalysis, including those involved in metal coordination (usually Mg²⁺)
The protein likely adopts a similar structure to other bacterial ispE enzymes, with specific variations that may relate to the adaptation of N. europaea to its ecological niche.
For successful expression of recombinant N. europaea ispE, researchers should consider the following methodological approaches:
E. coli expression systems: BL21(DE3) or similar strains are recommended due to their reduced protease activity and controllable expression via T7 promoter systems.
Expression vectors: pET series vectors containing N- or C-terminal affinity tags (His6, GST, or MBP) facilitate purification while potentially enhancing solubility.
Expression conditions:
Induce at OD₆₀₀ of 0.6-0.8 with 0.1-0.5 mM IPTG
Lower the temperature to 16-20°C after induction
Extend expression time to 16-20 hours
Supplement media with 5-10 mM MgCl₂ to stabilize the enzyme
Codon optimization: Consider codon optimization for E. coli if expression levels are low, as N. europaea and E. coli have different codon usage preferences.
Expression testing should include analysis of both soluble and insoluble fractions to determine optimal conditions for producing active enzyme.
A comprehensive purification strategy for recombinant N. europaea ispE should include:
Cell lysis buffer optimization:
50 mM Tris-HCl or HEPES, pH 7.5-8.0
100-300 mM NaCl
5-10% glycerol for stability
1-5 mM MgCl₂ (cofactor)
1-5 mM β-mercaptoethanol or DTT
Protease inhibitor cocktail
Affinity chromatography (primary purification):
For His-tagged constructs: Ni-NTA or TALON resin
Wash with increasing imidazole concentrations (20-40 mM)
Elute with 250-300 mM imidazole
Secondary purification:
Ion-exchange chromatography (IEX) using Q or SP columns depending on the isoelectric point
Size-exclusion chromatography (SEC) for highest purity and removal of aggregates
Quality control assessments:
SDS-PAGE for purity analysis (>95% for structural studies)
Western blot for identity confirmation
Dynamic light scattering (DLS) for monodispersity
Mass spectrometry for accurate mass determination
Circular dichroism (CD) for secondary structure confirmation
Enzyme activity should be assessed at each purification step to monitor retention of catalytic function.
The transcriptomic response of N. europaea to oxygen limitation provides important context for understanding ispE regulation. While the search results don't specifically mention ispE regulation under oxygen limitation, we can extrapolate based on related metabolic pathways in N. europaea:
Under oxygen-limited conditions, N. europaea experiences:
Metabolic reprogramming: A significant downregulation of carbon fixation genes, including RuBisCO-encoding genes (reduced by 2.3 to 6.3-fold) . This suggests a general reduction in biosynthetic activities that may extend to isoprenoid biosynthesis.
Energy allocation shifts: Polyphosphate accumulation increases under oxygen limitation (polyphosphate kinase transcription increases 2.1-fold) , indicating a reallocation of ATP resources. This may impact ATP-dependent reactions such as those catalyzed by ispE.
Respiratory adaptations: Increased expression of high-affinity terminal oxidases under oxygen limitation , suggesting prioritization of energy generation pathways over biosynthetic pathways.
To specifically study ispE regulation under oxygen limitation, researchers should:
Perform RT-qPCR targeting ispE during steady-state chemostat growth under both ammonia- and oxygen-limited conditions
Quantify protein levels using targeted proteomics approaches
Measure enzyme activity in cell extracts from cultures grown under different oxygen tensions
When designing kinetic assays for ispE activity, researchers should consider:
Coupled enzyme assays:
ADP production can be coupled to NADH oxidation via pyruvate kinase and lactate dehydrogenase
Monitor NADH decrease at 340 nm (ε = 6,220 M⁻¹cm⁻¹)
Reaction buffer: 50 mM HEPES or Tris-HCl (pH 7.5-8.0), 10 mM MgCl₂, 50 mM KCl
Direct phosphorylation assays:
Use [γ-³²P]ATP or [γ-³³P]ATP and measure radioactive phosphate incorporation
Separate products by thin-layer chromatography or HPLC
Quantify using phosphorimaging or scintillation counting
LC-MS/MS based assays:
Directly measure substrate depletion and product formation
Allows simultaneous monitoring of side reactions or contaminants
Higher sensitivity for detailed kinetic analyses
Optimization parameters:
Temperature range: 25-37°C (test at 5°C intervals)
pH range: 6.5-9.0 (test at 0.5 pH unit intervals)
Metal cofactors: Test Mg²⁺, Mn²⁺, Co²⁺ at 1-10 mM
ATP concentration: 0.1-5 mM
Substrate concentration: 0.1-2 mM
Site-directed mutagenesis represents a powerful approach for investigating the catalytic mechanism of N. europaea ispE. Based on conserved residues in GHMP kinases, the following experimental approach is recommended:
Target residue selection:
ATP-binding motifs (conserved glycine-rich regions)
Predicted catalytic residues (likely aspartate, lysine, or arginine residues)
Substrate-coordinating residues (typically polar amino acids)
Metal-coordinating residues (often aspartate or glutamate)
Mutation design strategy:
Conservative mutations: D→E, K→R to preserve charge
Non-conservative mutations: D→N, K→A to eliminate charge
Size variations: F→Y, S→A to test spatial requirements
Experimental workflow:
Use overlap extension PCR or commercial site-directed mutagenesis kits
Express and purify mutant proteins using identical conditions as wild-type
Compare expression levels, solubility, and thermal stability
Determine kinetic parameters (kcat, Km) for each mutant
Perform isothermal titration calorimetry to assess binding changes
Structural validation:
Circular dichroism to confirm proper folding
X-ray crystallography of key mutants with substrates/analogs
Molecular dynamics simulations to assess subtle structural changes
A systematic mutational analysis could generate a detailed model of the active site architecture and reaction mechanism, contributing to structure-based inhibitor design.
Comparative analysis of N. europaea ispE with homologs from other organisms can reveal evolutionary adaptations and functional differences. Consider the following methodological approach:
Sequence analysis:
Multiple sequence alignment with ispE from diverse bacteria, particularly comparing ammonia oxidizers with other bacterial groups
Phylogenetic tree construction to determine evolutionary relationships
Conservation analysis to identify N. europaea-specific variations
Structural comparative studies:
Homology modeling based on existing crystal structures
Molecular dynamics simulations under different conditions
Active site comparison focusing on substrate-binding residues
Functional comparison:
Express and purify homologs from representative organisms
Compare kinetic parameters under identical conditions
Test substrate specificity using substrate analogs
Evaluate thermal and pH stability profiles
Comparative data table for selected bacterial ispE enzymes:
| Organism | Size (aa) | Optimal pH | Optimal Temp (°C) | Km for substrate (μM) | kcat (s⁻¹) | Structure available |
|---|---|---|---|---|---|---|
| N. europaea | 278 | ? | ? | ? | ? | No |
| E. coli | ~283 | 7.5-8.0 | 30-37 | ~30-50 | ~1-5 | Yes (PDB: 1OJ4) |
| M. tuberculosis | ~283 | 7.0-7.5 | 37 | ~40-60 | ~0.5-2 | Yes |
| A. thaliana | ~365 | 8.0-8.5 | 25-30 | ~20-40 | ~0.5-2 | No |
Note: The table contains representative values based on general patterns in the literature. Exact values for N. europaea ispE would need to be experimentally determined.
Developing and studying inhibitors of N. europaea ispE requires a systematic approach:
Initial screening methods:
ATP-competitive inhibitor libraries (kinase inhibitor sets)
Fragment-based screening using differential scanning fluorimetry
In silico docking with homology models or crystal structures
Substrate analog design targeting unique features of the binding pocket
Secondary validation assays:
IC₅₀ determination using the optimized enzyme activity assay
Mechanism of inhibition studies (competitive, noncompetitive, uncompetitive)
Binding affinity measurements via isothermal titration calorimetry
Surface plasmon resonance for association/dissociation kinetics
Structure-activity relationship studies:
Systematic modification of lead compounds to improve potency
Focus on selectivity against human kinases
Address physicochemical properties for potential cellular penetration
Cellular validation:
Growth inhibition assays using N. europaea cultures
Metabolomic analysis to confirm on-target effects (accumulation of substrate)
Resistance studies to confirm mechanism of action
Potential inhibitor classes to explore:
Nucleotide analogs targeting the ATP binding site
Substrate competitive inhibitors mimicking 4-diphosphocytidyl-2C-methyl-D-erythritol
Allosteric inhibitors targeting unique regulatory sites
Covalent inhibitors targeting accessible cysteine residues
Integrating transcriptomic data can provide crucial insights into the metabolic context of ispE in N. europaea:
Co-expression network analysis:
Differential expression under varied conditions:
Integration with metabolomic data:
Correlate ispE expression levels with concentrations of isoprenoid precursors and products
Use isotope labeling to track carbon flux through the MEP pathway under different growth conditions
Develop a comprehensive model of isoprenoid biosynthesis regulation
Experimental validation approaches:
Construct reporter gene fusions to study promoter activity
Perform ChIP-seq to identify transcription factors binding to the ispE promoter
Create conditional knockdowns to assess the metabolic impact of reduced ispE activity
The transcriptomic study of N. europaea under oxygen limitation revealed significant downregulation of carbon fixation pathways , suggesting that isoprenoid biosynthesis might be similarly affected as part of a broader metabolic reprogramming during environmental stress.