Recombinant Nitrosomonas europaea Argininosuccinate synthase (argG) is a recombinant protein derived from the bacterium Nitrosomonas europaea. This enzyme plays a crucial role in the biosynthesis of arginine, an essential amino acid involved in various biological processes. Argininosuccinate synthase is part of the urea cycle and catalyzes the condensation of citrulline and aspartate to form argininosuccinate, a key intermediate in arginine production .
Product Code: CSB-EP767702NHH
Immunogen Species: Nitrosomonas europaea (strain ATCC 19718 / CIP 103999 / KCTC 2705 / NBRC 14298)
Purity: >85% (SDS-PAGE)
Sequence: The sequence begins with MDKVKKAVLA FSGGLDTSVI LKWLQDTYQC EVVTFTADIG QGEEIEPARA KAVQFGIREI FIEDLREEFV RDYVFPMFRA NTIYEGEYLL GTSIARPLIA KRQVEIAQQT GADAVSHGAT GKGNDQVRFE LGYYALQPDI RVIAPWREWD .
Argininosuccinate synthase is a urea cycle enzyme that catalyzes the condensation of citrulline and aspartate to form argininosuccinate. This reaction is crucial for the biosynthesis of arginine, which is involved in various physiological processes, including the synthesis of proteins, nitric oxide, and polyamines .
The catalytic reaction involves the formation of a citrullyl-AMP intermediate, which is stabilized by specific amino acid residues. The alpha-amino group of aspartate then attacks the imino carbon atom of this intermediate, producing AMP and argininosuccinate .
Nitrosomonas europaea is a well-studied ammonia-oxidizing bacterium that plays a crucial role in the nitrogen cycle. Its ability to oxidize ammonia to nitrite is essential for nitrogen cycling in ecosystems . The expression of genes involved in nitrogen metabolism, including those related to arginine synthesis, can be influenced by environmental conditions such as ammonia concentration .
Cusabio. Recombinant Nitrosomonas europaea Argininosuccinate synthase (argG) [CSB-EP767702NHH].
ASM Journals. Production and Excretion of Polyamines To Tolerate High Ammonia Concentrations in Ca. Nitrosocosmicus agrestis.
PMC. Functional Dissection of N-Acetylglutamate Synthase (ArgA) of Pseudomonas aeruginosa.
EMBL-EBI. Argininosuccinate synthetase.
PMC. Transcriptomic Response of Nitrosomonas europaea Transitioned from Aerobic to Oxygen-Limited Conditions.
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes. We will accommodate your request whenever possible.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its inclusion.
KEGG: neu:NE1437
STRING: 228410.NE1437
Argininosuccinate synthase (argG) in N. europaea catalyzes the ATP-dependent condensation of citrulline and aspartate to form argininosuccinate, a critical step in the arginine biosynthesis pathway. This enzyme is particularly important in N. europaea because:
It represents a key intersection between nitrogen assimilation and amino acid metabolism in this ammonia-oxidizing bacterium
The enzyme functions within the context of N. europaea's chemolithoautotrophic lifestyle, where energy is primarily derived from ammonia oxidation
Unlike heterotrophic organisms, N. europaea must coordinate argG activity with its unique energy metabolism that depends on ammonia availability
The utilization of aspartate by argG connects to the organism's limited capacity for amino acid uptake, as demonstrated in studies showing that L-aspartic acid can enhance growth and protein synthesis in N. europaea
Methodologically, researchers can confirm argG function in N. europaea through enzyme assays measuring the conversion of citrulline and aspartate to argininosuccinate, typically coupling this reaction to ATPase activity measurement or through direct quantification of argininosuccinate production.
N. europaea argG expression exhibits adaptive responses to various environmental stressors that affect the organism's core metabolism:
Oxygen limitation significantly alters N. europaea's transcriptional profile, with many metabolic genes showing differential expression
While specific argG transcription data under oxygen limitation is not directly reported, related amino acid metabolism genes show shifts in expression patterns during stress conditions
Similar to other metabolic genes, argG likely responds to the ATP availability fluctuations that occur when ammonia oxidation and ATP consumption become uncoupled
Experimental approaches to study argG expression include:
qRT-PCR to quantify argG mRNA levels under different growth conditions
Transcriptomics analysis comparing argG expression across various stressors
Reporter gene fusions to monitor argG promoter activity in vivo
| Environmental Condition | Expected argG Expression Response | Experimental Approach |
|---|---|---|
| Oxygen limitation | Likely altered based on related genes | Continuous culture with controlled DO levels, whole-genome transcriptomics |
| High nitrite exposure | Potentially upregulated to support stress response | Batch culture with added nitrite (250-300 mg-N/L), qRT-PCR analysis |
| Amino acid supplementation | May be downregulated with aspartate or glutamate addition | Growth with specific amino acids, enzyme activity assays |
| Ammonia limitation | Likely affected due to energy limitation | Chemostat cultures with controlled ammonia levels |
When cloning the N. europaea argG gene for recombinant expression, researchers should consider:
Genomic DNA extraction from N. europaea strain ATCC 19718, the standard reference strain with a sequenced genome
PCR amplification with high-fidelity polymerase and primers designed with appropriate restriction sites
Codon optimization may be necessary, as N. europaea has a different codon usage bias than common expression hosts
Vector selection should consider the need for controlled expression, as argG overexpression might be toxic
Methodological approach:
Extract genomic DNA using specialized protocols for Gram-negative bacteria
Design primers based on the annotated genome sequence, incorporating restriction sites compatible with expression vectors
PCR amplify using high-fidelity polymerase with optimized conditions for N. europaea's GC content
Clone into intermediate vectors for sequence verification before subcloning into expression vectors
Verify constructs by sequencing and restriction analysis
The optimal expression system for N. europaea argG depends on research objectives:
E. coli BL21(DE3) derivatives remain the first-choice system due to their high yield potential and compatibility with many expression vectors
For structural studies requiring high purity, specialized strains like E. coli Rosetta or ArcticExpress can improve folding
Cell-free expression systems may be valuable for rapid screening of functional variants
Expression optimization should address:
Temperature modulation (typically 16-25°C) to improve folding of N. europaea proteins
Inducer concentration titration to balance expression level with protein solubility
Media composition, potentially incorporating amino acids that enhance N. europaea growth, such as aspartate and glutamate
| Expression System | Advantages | Limitations | Best Applications |
|---|---|---|---|
| E. coli BL21(DE3) | High yield, simple cultivation | Potential inclusion bodies | Biochemical studies |
| E. coli Rosetta | Supplies rare tRNAs | Moderate yield | Structural studies |
| E. coli ArcticExpress | Enhanced folding at low temperature | Slower growth | Difficult-to-fold proteins |
| Cell-free systems | Rapid, toxicity-independent | Limited scale | Variant screening |
| Yeast systems | Eukaryotic processing | Complex media | Specialized studies |
Poor solubility of recombinant argG is a common challenge that can be addressed through:
Optimizing induction parameters: Lower IPTG concentrations (0.1-0.5 mM) and reduced temperatures (16-20°C) often improve solubility
Co-expression with molecular chaperones like GroEL/ES can assist proper folding
Fusion tags may enhance solubility, with maltose-binding protein (MBP) or SUMO tags often proving effective
Buffer optimization based on N. europaea's physiological conditions
Methodological approach to solubility screening:
Perform small-scale expression trials varying temperature, inducer concentration, and media composition
Analyze soluble vs. insoluble fractions by SDS-PAGE after cell lysis
Test multiple lysis buffers with different pH values (7.0-8.0), salt concentrations (100-500 mM NaCl), and additives
For persistent solubility issues, screen fusion tags (His, GST, MBP, SUMO) and co-expression with chaperones
Consider on-column refolding protocols if inclusion bodies cannot be avoided
Several factors can impact the enzymatic activity of recombinant argG:
Buffer composition: Optimal activity typically requires physiological pH (7.5-8.0) reflecting N. europaea's preferred environmental conditions
Metal ion requirements: Many argG enzymes require divalent cations (Mg²⁺) for optimal activity
Substrate availability: Both aspartate and citrulline must be present at appropriate concentrations
Redox environment: Activity may be sensitive to oxidizing conditions
Post-translational modifications may affect activity, though these are less common in bacterial systems
Optimizing for maximum activity requires:
Systematic screening of buffer conditions (pH 6.5-8.5)
Titration of divalent cations (Mg²⁺, Mn²⁺) at 1-10 mM concentrations
Substrate concentration optimization based on Km values
Addition of reducing agents (DTT, β-mercaptoethanol) to prevent oxidative inactivation
ATP concentration optimization, typically 1-5 mM
A multi-step purification strategy typically yields the best results for N. europaea argG:
Initial capture using affinity chromatography (His-tag or specific substrate affinity)
Intermediate purification by ion exchange chromatography
Polishing by size exclusion chromatography
Methodological approach:
Affinity chromatography: Immobilized metal affinity chromatography (IMAC) using Ni²⁺ or Co²⁺ resins for His-tagged constructs
Ion exchange chromatography: Based on argG's predicted isoelectric point, typically anion exchange (Q Sepharose) at pH 8.0
Size exclusion chromatography: Final polishing and buffer exchange, also providing information about oligomeric state
Optional tag removal using specific proteases if the tag affects activity
Buffer optimization is crucial throughout purification:
Incorporate stabilizing agents based on N. europaea's physiological environment
Consider including substrates or substrate analogs to stabilize the enzyme
Add reducing agents to prevent oxidative damage
Include glycerol (10-20%) to enhance stability during storage
Multiple complementary techniques should be employed to verify structural integrity:
Circular dichroism (CD) spectroscopy to assess secondary structure content
Differential scanning fluorimetry (DSF) to determine thermal stability and buffer optimization
Dynamic light scattering (DLS) to evaluate homogeneity and oligomeric state
Limited proteolysis to probe domain organization and folding quality
For advanced structural characterization:
X-ray crystallography remains the gold standard for high-resolution structure determination
Cryo-electron microscopy for structural analysis without crystallization
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to probe conformational dynamics
Small-angle X-ray scattering (SAXS) for solution-state structural information
| Technique | Information Provided | Sample Requirements | Advantages/Limitations |
|---|---|---|---|
| CD Spectroscopy | Secondary structure content | 0.1-0.5 mg/mL, 200-300 μL | Rapid, low sample consumption, limited resolution |
| DSF | Thermal stability, ligand binding | 0.1-0.2 mg/mL, 20-50 μL | High-throughput buffer screening, indirect structural information |
| DLS | Hydrodynamic radius, aggregation state | 0.5-1 mg/mL, 20-100 μL | Quick assessment of homogeneity, sensitive to aggregates |
| X-ray Crystallography | Atomic resolution structure | 5-10 mg/mL, 1-2 mL | Highest resolution, requires crystals |
| Cryo-EM | Near-atomic resolution structure | 1-5 mg/mL, 50-100 μL | No crystallization needed, requires specialized equipment |
The enzymatic activity of N. europaea argG can be measured using several approaches:
Coupled enzyme assays monitoring ADP production through pyruvate kinase and lactate dehydrogenase with NADH oxidation detection
Direct argininosuccinate product detection using HPLC or mass spectrometry
Radioactive assays using ¹⁴C-aspartate to track product formation
Optimal assay conditions typically include:
Temperature: 25-30°C, reflective of N. europaea's environmental preference
pH: 7.5-8.0, matching the organism's physiological pH
Buffer: 50 mM HEPES or Tris-HCl with 100-150 mM NaCl
Divalent cations: 5-10 mM MgCl₂ or MnCl₂
Substrates: Citrulline (1-5 mM), aspartate (1-5 mM), ATP (1-5 mM)
Reducing agent: 1-5 mM DTT to maintain enzymatic stability
For kinetic parameter determination:
Perform initial velocity measurements at various substrate concentrations
Plot data using appropriate kinetic models (Michaelis-Menten, Hill, etc.)
Determine Km, Vmax, kcat, and catalytic efficiency (kcat/Km)
Compare with argG enzymes from other organisms
N. europaea experiences several environmental stressors that may affect argG activity:
Oxygen limitation significantly alters N. europaea's metabolism and gene expression patterns
High nitrite concentrations (>250 mg-N/L) induce stress responses in N. europaea
Nutrient availability, particularly amino acids like aspartate and glutamate, influences growth and metabolism
To study these correlations:
Grow N. europaea under controlled conditions with specific stressors
Extract cellular proteins while preserving enzyme activity
Measure argG activity using standardized assays
Correlate activity with transcriptomic and metabolomic data
Investigate protein-protein interactions that may regulate argG under stress
A particularly relevant approach is studying argG activity in the context of oxygen limitation, as N. europaea shows significant transcriptional changes and metabolic adaptations under low oxygen conditions .
Understanding how argG integrates with N. europaea's unique metabolism requires specialized approaches:
Metabolic flux analysis using isotope-labeled substrates to track carbon and nitrogen flow
Reconstitution of connected enzymatic pathways in vitro
Protein-protein interaction studies to identify regulatory partners
Methodological approaches include:
Isotope labeling experiments using ¹³C-labeled bicarbonate or ¹⁵N-labeled ammonia
Metabolomics analysis of pathway intermediates under various conditions
Pull-down assays and co-immunoprecipitation to identify interaction partners
In vitro reconstitution of connected pathways with purified enzymes
Systems biology approaches integrating transcriptomics, proteomics, and metabolomics data
| Approach | Information Obtained | Technical Considerations |
|---|---|---|
| Isotope Labeling | Metabolic flux distribution | Requires mass spectrometry, specialized analysis software |
| Metabolomics | Pathway intermediate levels | Sample preparation critical, requires rapid quenching |
| Protein-Protein Interactions | Regulatory connections | May require cross-linking for transient interactions |
| Pathway Reconstitution | Direct enzyme connections | Multiple purified enzymes needed, complex assay development |
The interaction between argG and N. europaea's central ammonia oxidation pathway presents an intriguing research area:
N. europaea derives energy from ammonia oxidation via ammonia monooxygenase (AMO) and hydroxylamine dehydrogenase (HAO)
ArgG requires ATP generated from this energy metabolism, creating a dependency relationship
Aspartate utilization by argG may interface with amino acid uptake pathways, shown to affect N. europaea growth
Under stress conditions like oxygen limitation, the transcription of central metabolism genes changes significantly , likely affecting argG function
Research approaches for studying these interactions:
Metabolic modeling to predict flux distributions between energy generation and arginine biosynthesis
Comparative transcriptomics to correlate argG expression with ammonia oxidation genes under various conditions
ATP availability assays to determine how energy status affects argG activity
Isotope labeling to track nitrogen flow between ammonia oxidation and arginine biosynthesis
The integration likely involves regulatory mechanisms sensing energy status and nitrogen availability, connecting argG to N. europaea's adaptation to changing environmental conditions.
Oxygen limitation represents a significant stress for obligate aerobe N. europaea, with transcriptomic data revealing:
During oxygen-limited growth, N. europaea shows reduced growth yield and non-stoichiometric ammonia-to-nitrite conversion
Many metabolic genes show differential expression under oxygen limitation, including those involved in energy conservation and biosynthetic pathways
The data suggests increased polyphosphate accumulation during O₂-limited growth, which may serve as an energy reserve affecting ATP-dependent processes like the argG reaction
While specific argG transcription under oxygen limitation isn't directly reported in the available data, related observations suggest:
Similar ATP-dependent biosynthetic enzymes show altered expression under oxygen limitation
Changes in energy conservation pathways likely impact all ATP-requiring enzymes, including argG
The differential regulation of amino acid metabolism genes suggests argG would also be affected
Research approaches:
qRT-PCR specifically targeting argG under controlled oxygen conditions
Enzyme activity assays comparing argG function in extracts from oxygen-limited vs. oxygen-sufficient cultures
Metabolomics targeting arginine pathway intermediates under varying oxygen conditions
Reporter gene fusions to monitor argG promoter activity in vivo during oxygen transitions
N. europaea produces nitrite as the end product of ammonia oxidation, and high nitrite concentrations can become toxic to the cells:
N. europaea has evolved specific mechanisms to deal with nitrite toxicity, including transcriptional responses
High nitrite concentrations (280 mg-N/L) cause elevated expression of genes involved in nitrite detoxification, such as nirK and norB
The connection between argG and nitrite detoxification pathways may involve shared regulatory elements or metabolic interactions
Research approaches to investigate this relationship:
Comparative growth experiments with wild-type and argG-modified strains under high nitrite conditions
Transcriptomic analysis comparing argG expression with nitrite response genes under nitrite stress
Metabolite profiling to identify changes in arginine pathway intermediates during nitrite exposure
Investigation of potential regulatory crossover between nitrite response and arginine biosynthesis pathways
Understanding this relationship could provide insights into how N. europaea coordinates biosynthetic processes with stress responses in its specialized ecological niche.
Creating targeted genetic modifications in N. europaea requires specialized approaches:
Homologous recombination-based methods using suicide vectors that cannot replicate in N. europaea
Selection systems appropriate for this ammonia oxidizer, which has different antibiotic sensitivities than model organisms
Careful control of growth conditions during transformation and selection
Methodological approach:
Design homologous flanking regions (1-2 kb) surrounding the argG target locus
Clone these regions into a suicide vector containing appropriate selection markers
Transform N. europaea using electroporation optimized for this organism
Select for single crossovers using appropriate antibiotics
Counter-select for double crossovers to generate clean deletions or replacements
Verify mutations by PCR, sequencing, and phenotypic analysis
Alternatives include:
CRISPR-Cas9 systems adapted for N. europaea
Transposon mutagenesis for random insertion libraries that can be screened for argG disruptions
Antisense RNA approaches for partial knockdown when complete knockouts are lethal
Comprehensive phenotypic analysis of argG mutants should include:
Growth characteristics under various conditions (different ammonia concentrations, oxygen levels, etc.)
Ammonia oxidation rates measured by oxygen uptake or nitrite production
Arginine auxotrophy testing with supplementation studies
Stress tolerance profiling (nitrite, oxygen limitation, pH stress)
Transcriptomic and metabolomic comparisons with wild-type
Methodological approaches:
Growth curve analysis in minimal and supplemented media
Respirometry to measure oxygen consumption rates
Colorimetric assays for nitrite production
Metabolite profiling using targeted LC-MS/MS for arginine pathway intermediates
RNA-Seq to identify compensatory transcriptional responses
Survival assays under various stress conditions
For conditional mutants or partial knockdowns:
Titrate inducer/repressor concentrations to vary expression levels
Correlate phenotypic effects with argG activity levels
Identify threshold levels required for normal growth and metabolism
Complementation studies are essential to confirm that observed phenotypes result directly from argG modification:
Reintroduction of wild-type argG at a neutral chromosomal site or on a stable plasmid
Expression under native or controlled promoters to ensure appropriate levels
Inclusion of tagged versions for localization and interaction studies
Methodological approach:
Clone wild-type argG with its native promoter and ribosome binding site
Introduce variations: point mutations, heterologous argG genes, tagged versions
Transform into argG mutant strains
Verify expression by qRT-PCR, Western blotting, or enzyme activity assays
Assess restoration of growth, ammonia oxidation, and arginine synthesis
Compare complementation efficiency across different constructs and expression levels
| Complementation Approach | Advantages | Limitations | Best Applications |
|---|---|---|---|
| Chromosomal integration | Stable, single-copy expression | Complex construction | Long-term studies |
| Plasmid-based | Easier construction, multiple copies | Stability issues, selection needed | Initial characterization |
| Inducible systems | Controlled expression timing and level | Leaky expression, complex regulation | Dosage studies |
| Heterologous argG | Functional comparisons across species | May not fully complement | Evolutionary studies |
Systems biology offers powerful approaches to understand argG in the context of N. europaea's complete metabolism:
Methodological approach:
Construct or refine existing N. europaea metabolic models to accurately represent argG reactions
Constrain models using experimental data on growth rates, substrate utilization, and product formation
Perform in silico gene knockouts or modifications to predict system-wide effects
Validate predictions with targeted experiments
Iteratively refine models based on experimental results
Advanced applications include:
Identification of potential synthetic lethal interactions with argG
Prediction of metabolic bottlenecks that could be targeted for strain improvement
Understanding how argG flux affects global nitrogen and carbon distribution
Understanding the structural adaptations of N. europaea argG provides insights into its evolution:
Comparative structural analysis between N. europaea argG and homologs from other organisms
Identification of unique regions that may interface with N. europaea-specific regulatory partners
Analysis of substrate binding sites and catalytic residues for specialized features
Research approaches:
Homology modeling based on crystallized argG structures from related organisms
X-ray crystallography or cryo-EM studies of purified N. europaea argG
Site-directed mutagenesis targeting unique residues
Molecular dynamics simulations to analyze structural flexibility and substrate interactions
Evolutionary analysis to identify positively selected residues
Structural insights could reveal:
Adaptations to N. europaea's specific intracellular environment (pH, ion concentrations)
Regulatory interfaces unique to this ammonia oxidizer
Potential allosteric regulation sites connecting argG to nitrogen metabolism
Modern high-throughput technologies offer new avenues to study argG regulation:
ChIP-Seq to identify transcription factors binding to the argG promoter
RNA-Seq across multiple conditions to create regulatory networks
Proteomics approaches to identify post-translational modifications
High-throughput mutagenesis combined with phenotypic screening
Methodological approach:
Generate reporter strains with fluorescent proteins driven by the argG promoter
Screen libraries of environmental conditions or chemical perturbagens
Perform transposon sequencing (Tn-Seq) to identify genetic interactions
Use CRISPR interference screens targeting potential regulators
Apply machine learning to integrate diverse datasets and predict regulatory relationships
These approaches can reveal:
Condition-specific regulation of argG expression
Novel regulatory factors not previously associated with arginine metabolism
Integration of argG regulation with global stress responses
Potential applications for metabolic engineering of N. europaea
Despite advances in understanding N. europaea metabolism, several critical questions about argG remain:
The precise regulatory mechanisms connecting argG to ammonia oxidation and energy generation
The role of argG in environmental adaptation and stress responses
Structural adaptations that may distinguish N. europaea argG from homologs in other organisms
The potential for argG manipulation to improve N. europaea performance in biotechnological applications
Future research directions should focus on:
Integrating argG studies into systems-level analyses of N. europaea metabolism
Applying advanced structural biology techniques to elucidate unique features
Investigating regulatory networks connecting argG to environmental sensing
Exploring the evolutionary history of argG in ammonia-oxidizing bacteria
Developing genetic tools specifically optimized for studying arginine metabolism in N. europaea