Note: We will prioritize shipping the format currently in stock. If you have specific format requirements, please indicate them during order placement. We will accommodate your request to the best of our ability.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested. Please contact us in advance; additional fees will apply.
The tag type will be determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Converts GTP to 7,8-dihydroneopterin triphosphate.
KEGG: neu:NE1163
STRING: 228410.NE1163
GTP cyclohydrolase folE2 is an enzyme involved in the tetrahydrobiopterin biosynthetic pathway, catalyzing the conversion of GTP to dihydroneopterin triphosphate. In Nitrosomonas europaea, this enzyme plays a critical role in folate metabolism and potentially in nitrogen metabolism pathways. As a member of the GTP cyclohydrolase family (EC 3.5.4.16), folE2 is structurally distinct from folE1 but performs related catalytic functions in pteridine biosynthesis .
Nitrosomonas europaea folE2 is a full-length protein consisting of 268 amino acids. The recombinant form expressed for research purposes maintains the complete amino acid sequence from position 1-268. The protein has the following key properties:
Molecular weight: Approximately 20.2 kDa
Amino acid sequence: MNKQIDLPIADVQGSLDTRHIAIDRVGIKAIVAIRHPVVVADKGGGSQHTVAQFNMYVNLPHNFKGTHMSRFVEILNSHEREISVESFEEILRSMVSRLESDSGHIEMAFPYFINKSAPVSGVKSLLDYEVTFIGEIKHGNQYSFTMKVIVPVTSLCPCSKISDYGAHNQRSHVTISVRTNSFIWIEDIIRIAEEQASCELYGLLKRPDEKYVOIRTRAYLNNPKFVEDIVRDVIAEVLNHDDRIDAYIVESENESIHNHSAYALIERODKRIR
Nitrosomonas europaea folE2 shares structural and functional homology with similar enzymes found across diverse bacterial genera. Comparative analysis shows approximately 56.7% sequence identity with human GTP cyclohydrolase 1. This degree of conservation suggests essential metabolic functions. When examining folE2 from Pseudomonas aeruginosa, it belongs to a large ortholog group (POG003716) with 533 members, indicating wide distribution across bacterial species . The presence of folE2 across 471 genera further emphasizes its evolutionary significance and common occurrence in both pathogenic and non-pathogenic bacterial strains .
Multiple expression systems have been successfully employed for the production of recombinant Nitrosomonas europaea folE2, each with distinct advantages:
| Expression System | Product Code | Advantages | Considerations |
|---|---|---|---|
| E. coli | CSB-EP774299NHH | High yield, cost-effective, rapid growth | May form inclusion bodies requiring refolding |
| Yeast | CSB-YP774299NHH | Post-translational modifications, secreted expression | Longer production time, complex media |
| Baculovirus | CSB-BP774299NHH | Native-like folding, high expression levels | Technical complexity, higher cost |
| Mammalian cell | CSB-MP774299NHH | Superior post-translational modifications | Highest cost, slowest production |
The E. coli expression system represents the most commonly used approach due to its efficiency and cost-effectiveness for basic research applications .
For optimal purification of functional recombinant folE2, a multi-step protocol is recommended:
Initial clarification: Centrifugation of lysed cells (10,000g, 30 minutes, 4°C)
Capture step: Immobilized metal affinity chromatography (IMAC) using Ni-NTA for His-tagged folE2
Intermediate purification: Ion exchange chromatography (recommended buffer: 20 mM Tris-HCl, pH 8.0)
Polishing step: Size exclusion chromatography
Quality control: SDS-PAGE analysis to confirm >85% purity
For biotinylated variants (e.g., CSB-EP774299NHH-B), the E. coli biotin ligase (BirA) catalyzes the covalent attachment of biotin to the AviTag peptide with high specificity, enabling additional purification options via avidin-based affinity chromatography .
Proper reconstitution is critical for maintaining enzymatic activity. The recommended protocol includes:
Brief centrifugation of the vial prior to opening to ensure all lyophilized material is at the bottom
Reconstitution in deionized sterile water to a concentration of 0.1-1.0 mg/mL
Addition of glycerol to a final concentration of 5-50% to enhance stability
Gentle mixing by slow inversion rather than vortexing to prevent protein denaturation
Storage at -20°C for medium-term use or -80°C for long-term storage
Several analytical methods can quantify the enzymatic activity of recombinant folE2:
Spectrophotometric assay: Monitoring the conversion of GTP to dihydroneopterin triphosphate at 330 nm
HPLC analysis: Quantifying reaction products using reverse-phase HPLC with UV detection
Coupled enzyme assay: Using alkaline phosphatase to remove phosphates from the product, followed by iodine oxidation and fluorescence detection
Mass spectrometry: LC-MS/MS analysis for precise quantification of reaction products
The selection of the appropriate assay depends on available instrumentation and required sensitivity. For high-throughput screening applications, the fluorescence-based coupled assay offers the best combination of sensitivity and throughput .
Nitrosomonas europaea is an obligate ammonia-oxidizing bacterium, and the relationship between folE2 activity and ammonia oxidation represents an important area of investigation. Current research suggests:
Folate metabolism, influenced by folE2 activity, may impact cellular energy production required for ammonia oxidation
Changes in environmental ammonia concentrations (e.g., 50 mM ammonium media) can affect metabolic pathways involving folE2
The production of nitrite, a primary indicator of ammonia oxidation activity, may correlate with folE2 expression levels
The function of folE2 in Nitrosomonas europaea can be significantly influenced by environmental conditions:
| Environmental Factor | Effect on folE2 | Research Implications |
|---|---|---|
| pH changes | Activity modulation | Study optimal pH range (typically 7.0-8.0) |
| Oxygen availability | Expression regulation | Analyze transcriptional changes under varying O₂ |
| Ammonia concentration | Potential activity correlation | Investigate dose-dependent relationships |
| Heavy metal exposure | Possible inhibition | Examine environmental toxicology impacts |
Research methodologies should incorporate careful control of these variables when studying folE2 function, particularly when extrapolating laboratory findings to environmental contexts .
The catalytic mechanism of folE2 depends on several key structural features:
Active site residues: Critical amino acids involved in substrate binding and catalysis
Metal ion coordination: Potential requirement for zinc or other divalent cations
Protein folding dynamics: Conformational changes during catalysis
Oligomerization state: Functional unit may be monomeric, dimeric, or higher-order structures
X-ray crystallographic studies of related folE2 proteins have revealed insights into inhibitor binding. For example, research on folE2 from Burkholderia thailandensis demonstrated that dehydrocostus lactone (DHL) functions as a mechanism-based inhibitor, with crystal structures capturing the covalently inhibited enzyme .
Comparative analysis between Nitrosomonas europaea folE2 and folE2 from pathogenic bacteria reveals important considerations for antimicrobial research:
FolE2 has been identified as conditionally essential in Burkholderia thailandensis and Burkholderia pseudomallei in the presence of subinhibitory doses of trimethoprim
Chemical synthetic lethality approaches have identified inhibitors like dehydrocostus lactone (DHL), parthenolide, and β-lapachone that form lethal combinations with low-dose trimethoprim
The concept of targeting folE2 may represent a promising strategy for developing new antimicrobial combinations against specific pathogens
Structural similarities between folE2 proteins across bacterial species suggest potential for broad-spectrum inhibitor development
Researchers studying Nitrosomonas europaea folE2 can leverage these insights for comparative structural analysis and inhibitor design, potentially contributing to antimicrobial discovery efforts .
Robust experimental design for folE2 enzymatic studies requires multiple controls:
Negative enzyme control: Reaction mixture without folE2 to establish baseline
Denatured enzyme control: Heat-inactivated folE2 to confirm activity is enzyme-dependent
Substrate saturation control: Varying GTP concentrations to ensure non-limiting conditions
Buffer composition control: Testing activity in different buffer systems to optimize conditions
Metal ion dependency control: Addition/chelation of potential cofactors
Thermal stability control: Activity measurements at different temperatures
These controls help ensure experimental reproducibility and valid interpretation of results when characterizing folE2 enzymatic properties .
When screening for folE2 inhibitors, consider the following experimental design principles:
Primary screening: High-throughput fluorescence-based assays to identify initial hits
Secondary validation: Orthogonal assays using different detection methods to confirm true positives
Mechanism studies: Kinetic analysis to determine inhibition type (competitive, non-competitive, uncompetitive)
Structure-activity relationship analysis: Testing structural analogs to identify key pharmacophores
X-ray crystallography: Obtaining structures of enzyme-inhibitor complexes for rational design
Cellular assays: Testing inhibitor effects in appropriate bacterial models
Recent research exemplifies this approach, where researchers identified dehydrocostus lactone as a mechanism-based inhibitor of folE2, confirmed through X-ray crystallography of the covalently inhibited enzyme .
Several methodologies can effectively investigate folE2 expression regulation:
| Methodology | Application | Advantages | Limitations |
|---|---|---|---|
| qRT-PCR | Quantitative mRNA levels | High sensitivity, specific | Limited to transcript level |
| Western blotting | Protein expression | Direct protein quantification | Antibody availability |
| Reporter gene assays | Promoter activity | Real-time monitoring possible | Requires genetic modification |
| RNA-seq | Transcriptome-wide context | Comprehensive gene expression | Complex data analysis |
| ChIP-seq | Transcription factor binding | Identifies regulatory elements | Technical complexity |
| CRISPR interference | Gene knockdown | Targeted repression | Potential off-target effects |
Combining multiple approaches provides the most comprehensive understanding of folE2 expression regulation under various environmental and experimental conditions .
Comparative analysis reveals both similarities and differences among folE2 proteins across bacterial species:
| Aspect | Nitrosomonas europaea folE2 | Pseudomonas aeruginosa folE2 | Burkholderia thailandensis folE2 |
|---|---|---|---|
| Length | 268 amino acids | Similar (full sequence not provided) | Similar (full sequence not provided) |
| Function | GTP cyclohydrolase activity | GTP cyclohydrolase I precursor | Similar enzymatic function |
| Essential nature | Not fully characterized | Common in both pathogenic and non-pathogenic strains | Conditionally essential with trimethoprim |
| Inhibitor susceptibility | Not fully characterized | Not reported in provided data | Inhibited by DHL, parthenolide, β-lapachone |
| Evolutionary conservation | Part of large ortholog group | Part of POG003716 with 533 members | Similar conservation pattern |
This comparative approach provides valuable insights into conserved features that may be essential for enzymatic function versus species-specific characteristics that could be exploited for selective targeting .
The folE1 and folE2 enzyme families represent distinct types of GTP cyclohydrolases with important differences:
Evolutionary origin: folE2 evolved independently from folE1, representing convergent evolution toward similar catalytic functions
Structural differences: Different protein folds despite catalyzing similar reactions
Metal ion requirements: Potentially different dependencies on zinc or other divalent cations
Substrate specificity: May show different affinities for GTP or substrate analogs
Inhibition profiles: Typically respond differently to known inhibitors
Distribution across species: Different patterns of presence across bacterial phyla
These differences have significant implications for researchers studying folE2, particularly when extrapolating findings from folE1 studies or when designing selective inhibitors .
Researchers frequently encounter several technical challenges when working with recombinant folE2:
Protein solubility issues: Formation of inclusion bodies in E. coli expression systems
Solution: Optimize expression temperature (typically lowering to 18-25°C), use solubility-enhancing tags, or employ refolding protocols
Enzymatic activity loss during purification:
Solution: Include stabilizing agents (glycerol, reducing agents), minimize purification steps, and maintain cold temperature throughout
Protein aggregation during storage:
Solution: Add glycerol (5-50%), aliquot to avoid freeze-thaw cycles, and store at -80°C
Low expression yields:
Solution: Optimize codon usage for expression host, test different promoter systems, or switch to alternative expression hosts
Protein degradation:
Optimizing enzymatic assays for folE2 requires attention to several key factors:
Buffer composition: Test multiple buffer systems (Tris, HEPES, phosphate) at various pH values (typically 7.0-8.5)
Cofactor requirements: Systematically test divalent cations (Mg²⁺, Zn²⁺, Mn²⁺) at different concentrations
Substrate concentration optimization: Determine Km value and use 2-5 times Km for routine assays
Temperature optimization: Test activity at different temperatures (typically 25-37°C)
Detection method sensitivity: For spectrophotometric methods, ensure measurements are within the linear range; for fluorescence-based methods, control for background fluorescence
Reaction time course: Establish linear range of product formation to ensure measurements are taken during initial velocity phase
Several emerging research directions for folE2 in Nitrosomonas europaea hold particular promise:
Role in nitrogen cycling: Investigating how folE2 activity influences ammonia oxidation rates in environmental contexts
Biosensor development: Exploring folE2-based biosensors for environmental monitoring applications
Metagenomic analysis: Examining folE2 distribution and variants across diverse environmental microbiomes
Climate change impacts: Studying how altered environmental conditions affect folE2 expression and function
Bioremediation applications: Investigating potential roles in systems for nitrogen removal from contaminated sites
These research directions connect fundamental enzyme biochemistry to broader ecological and environmental applications, potentially addressing significant challenges in environmental microbiology and biotechnology .
Comparative genomics offers powerful approaches to elucidate folE2 evolution and function:
Phylogenetic analysis: Constructing evolutionary trees based on folE2 sequences across diverse species
Synteny analysis: Examining conservation of genomic context around folE2 genes
Selection pressure analysis: Calculating dN/dS ratios to identify regions under positive or purifying selection
Structural homology modeling: Predicting folE2 structures in understudied organisms based on resolved structures
Horizontal gene transfer investigation: Identifying potential HGT events in folE2 evolutionary history
The widespread distribution of folE2 across 471 genera and its presence in a large ortholog group (POG003716) with 533 members provides a rich dataset for such comparative approaches .