Recombinant Pseudomonas putida N-acetyl-gamma-glutamyl-phosphate reductase 1 (argC1) refers to a genetically engineered form of the enzyme N-acetyl-gamma-glutamyl-phosphate reductase (NAGPR), which is originally derived from the bacterium Pseudomonas putida. NAGPR, encoded by the gene argC, is a crucial enzyme in the arginine biosynthetic pathway, catalyzing the reduction of N-acetyl-L-glutamate-5-semialdehyde to N-acetyl-L-ornithine .
Pseudomonas putida: P. putida is a Gram-negative bacterium known for its metabolic versatility and ability to tolerate various environmental conditions, making it a valuable host for producing natural products and other biotechnological applications .
Recombinant Production: The "recombinant" aspect indicates that the argC1 gene has been isolated, modified, and reintroduced into P. putida or another host organism to enhance enzyme production or study its properties . This often involves using techniques of genetic engineering to optimize the expression of the enzyme .
N-acetyl-gamma-glutamyl-phosphate reductase 1 (argC1): This enzyme is involved in the sixth step of arginine biosynthesis . Specifically, NAGPR catalyzes the NADPH-dependent reduction of N-acetyl-L-glutamate 5-semialdehyde to N-acetyl-L-ornithine .
Arginine is an essential amino acid involved in various cellular processes, including protein synthesis, nitrogen transport, and the production of nitric oxide . The argC1 gene product, N-acetyl-gamma-glutamyl-phosphate reductase, plays a vital role in the synthesis of arginine. The enzyme catalyzes a key step in the arginine biosynthetic pathway, ensuring the continuous production of arginine .
Metabolic Engineering: Recombinant P. putida argC1 can be used to enhance arginine production in P. putida strains. Overexpression of argC1, along with other enzymes in the arginine pathway, can lead to increased arginine biosynthesis, which has implications in various industrial and biotechnological applications .
Production of Natural Products: P. putida strains are engineered to produce valuable natural products, including rhamnolipids, terpenoids, polyketides, and non-ribosomal peptides . Engineered strains of P. putida exhibited high production of both compounds after a single chromosomal integration of the corresponding BGC .
Bioremediation: P. putida's ability to tolerate xenobiotics makes it useful in bioremediation. Recombinant P. putida strains expressing specific enzymes can degrade environmental pollutants. The esterase enzyme derived from P. putida can perform stereoselective hydrolysis, which is valuable in producing optically pure compounds .
Enzyme Optimization and Study: The recombinant form of argC1 allows researchers to produce and purify the enzyme in large quantities for structural and functional studies. These studies help elucidate the enzyme's mechanism, kinetics, and regulation, providing insights into its role in cellular metabolism .
Whole-Cell Biocatalysts: P. putida can be used as a host for the autotransporter-mediated surface display of enzymes, resulting in whole-cell biocatalysts with recombinant functionalities on their cell envelope .
Several techniques are employed to create recombinant P. putida strains:
Gene Cloning and Expression: The argC1 gene is isolated and cloned into a plasmid vector. This vector is then introduced into P. putida, where the gene is expressed under the control of a suitable promoter. Various promoters and expression systems can be used to optimize the production of NAGPR .
Chromosomal Integration: Instead of using plasmids, the argC1 gene can be integrated directly into the P. putida chromosome. This provides a more stable genetic modification and can lead to more consistent enzyme production .
Mutagenesis: Random or site-directed mutagenesis can be used to modify the argC1 gene, potentially improving enzyme activity, stability, or other desirable properties. These modified genes are then expressed in P. putida to assess their effects .
While P. putida is a promising host for recombinant protein production, there are challenges:
Optimization of Expression: Achieving high levels of recombinant protein expression can be challenging. Factors such as codon usage, mRNA stability, and protein folding can affect the final yield of active enzyme .
Metabolic Load: Overexpression of recombinant proteins can place a metabolic burden on the host cell, affecting growth and viability .
Strain Stability: Maintaining the stability of recombinant strains during long-term cultivation can be difficult, as the introduced genes may be lost or inactivated .
Future research directions include:
Advanced Genetic Tools: Developing more sophisticated genetic tools for P. putida, such as CRISPR-Cas systems and improved promoter libraries, can enhance the precision and efficiency of genetic engineering .
Systems Biology Approaches: Using systems biology approaches to understand and optimize the metabolic network of P. putida can help to minimize the metabolic burden of recombinant protein production and maximize yields .
Bioreactor Design: Optimizing bioreactor conditions for recombinant P. putida cultivation, including nutrient supply, oxygen levels, and pH control, can improve cell growth and protein production .
Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type will be determined during production. Please inform us of any specific tag requirements for preferential development.
Catalyzes the NADPH-dependent reduction of N-acetyl-5-glutamyl phosphate to N-acetyl-L-glutamate 5-semialdehyde.
KEGG: ppu:PP_0432
STRING: 160488.PP_0432
N-acetyl-gamma-glutamyl-phosphate reductase (AGPR) is composed of alpha/beta and alpha+beta domains with the active site located in the cleft between these domains. Similar to the Mycobacterium tuberculosis AGPR structure, P. putida AGPR likely binds the cofactor NADP+ in this interdomain cleft. The enzyme adopts a specific conformation upon NADP+ binding, with a notable movement (approximately 5 Å) in a loop region (analogous to the Leu88-His92 loop in MtbAGPR) to create a hydrophobic pocket that confines the adenine moiety of the cofactor . For experimental structure determination, researchers typically employ X-ray crystallography at resolutions better than 2.5 Å to accurately model the active site architecture.
P. putida argC1 catalyzes the NADPH-dependent reductive dephosphorylation of N-acetyl-gamma-glutamyl-phosphate to N-acetylglutamate-gamma-semialdehyde, a critical step in the arginine biosynthetic pathway. The catalytic mechanism likely involves several key residues forming a catalytic triad. Based on structural studies of similar AGPRs, a cysteine residue (equivalent to Cys158 in MtbAGPR) likely performs the nucleophilic attack on the substrate's gamma-carboxyl group. A histidine residue (equivalent to His219) acts as a general base, accepting a proton from the catalytic cysteine, while another histidine (equivalent to His217) positions the substrate through hydrogen bonding . Additionally, an arginine residue (equivalent to Arg114) forms an ion pair with the substrate's phosphate group for optimal positioning, and a glutamate residue stabilizes the positive charge that develops on the catalytic histidine during the reaction .
Within the arginine biosynthetic pathway, argC1 catalyzes the second step, converting N-acetyl-gamma-glutamyl-phosphate to N-acetylglutamate-gamma-semialdehyde. This reaction follows the acetylation of glutamate and precedes the transamination step that forms N-acetylornithine. The pathway is essential for arginine biosynthesis in many microorganisms and plants, including P. putida . The functional importance of this pathway makes it an attractive target for antimicrobial research, as human cells lack this biosynthetic route. Researchers investigating this pathway typically employ metabolic flux analysis using isotope-labeled precursors to measure pathway activity under various conditions.
While specific data for argC1 expression is limited, lessons from recombinant protein production in P. putida suggest the following optimized conditions:
For optimal results, researchers should monitor growth curves and protein expression levels to determine strain-specific optimal harvest times. Additionally, supplementing the growth medium with cofactors such as NADP+ may improve enzyme activity in the recombinant system.
For stable expression of argC1 in P. putida, chromosomal integration offers advantages over plasmid-based expression. Effective strategies include:
Verification of argC1 expression and functionality can be performed through multiple complementary approaches:
Enzyme activity assays: Measure the NADPH-dependent reduction of N-acetyl-gamma-glutamyl-phosphate to N-acetylglutamate-gamma-semialdehyde spectrophotometrically by monitoring NADPH oxidation at 340 nm.
Western blot analysis: Using antibodies against argC1 or epitope tags if the recombinant enzyme is tagged.
RT-qPCR: Quantify argC1 mRNA levels to confirm transcription.
Growth complementation: Test whether the recombinant strain can grow in minimal medium without arginine, which would confirm a functional arginine biosynthetic pathway.
Insertion site verification: Use plasmid rescue techniques and sequencing to identify the exact chromosomal location of the integrated argC1 gene, similar to the approach described for prodigiosin gene integration verification .
A comprehensive purification protocol for recombinant P. putida argC1 typically includes:
Cell lysis: Sonication or high-pressure homogenization in a buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10% glycerol, and 1 mM DTT.
Initial clarification: Centrifugation at 15,000 × g for 30 minutes to remove cell debris.
Affinity chromatography: If the recombinant protein contains an affinity tag (His-tag, GST-tag), use the appropriate affinity resin. For His-tagged proteins, use Ni-NTA chromatography with imidazole gradient elution.
Ion-exchange chromatography: Further purification using anion exchange chromatography (e.g., Q Sepharose) with a NaCl gradient.
Size-exclusion chromatography: Final polishing step using a Superdex 200 column to obtain homogeneous enzyme preparations.
Quality control: SDS-PAGE analysis to confirm purity, and enzyme activity assays to confirm functionality. Mass spectrometry can be used to verify protein identity.
Throughout purification, it's crucial to include NADP+ in the buffers to stabilize the enzyme structure, as binding of this cofactor induces conformational changes that may enhance protein stability .
Key kinetic parameters for argC1 and recommended measurement methods include:
| Parameter | Typical Range | Measurement Method |
|---|---|---|
| Km for N-acetyl-gamma-glutamyl-phosphate | 50-500 μM | Vary substrate concentration, monitor NADPH oxidation rate |
| Km for NADPH | 10-100 μM | Vary NADPH concentration at saturating substrate levels |
| kcat | 1-20 s^-1 | Calculate from Vmax and enzyme concentration |
| pH optimum | 7.0-8.5 | Measure activity across pH range using appropriate buffers |
| Temperature optimum | 25-37°C | Measure activity across temperature range |
| Inhibition by product | Ki = 0.1-1 mM | Product inhibition studies with varying inhibitor concentrations |
For accurate measurements, researchers should:
Use freshly prepared substrate solutions
Ensure linear reaction rates during measurements
Control temperature precisely
Account for any background NADPH oxidation
Use appropriate curve-fitting methods (e.g., non-linear regression to Michaelis-Menten equation)
Analyzing conformational changes in argC1 upon NADP+ binding requires several biophysical techniques:
When interpreting these data, researchers should focus on the interdomain cleft and potential loop movements that facilitate cofactor binding, as these are likely critical for the enzyme's catalytic function .
Site-directed mutagenesis of key catalytic residues can elucidate their specific roles in the enzymatic mechanism of argC1. Based on structural homology with MtbAGPR, researchers could target residues equivalent to Cys158, His217, His219, Arg114, and Glu222 . A systematic mutagenesis approach might include:
Cysteine to serine mutation: To test the role of the nucleophilic thiol group while maintaining similar sterics.
Histidine to alanine or asparagine: To assess the importance of the imidazole group in substrate positioning and proton transfer.
Arginine to lysine or glutamine: To investigate the specificity of substrate phosphate binding.
Glutamate to glutamine or aspartate: To examine the role in stabilizing the charged histidine.
Each mutant should be characterized through:
Expected outcomes might include complete loss of activity for mutations in the nucleophilic cysteine, substantial reduction in activity with histidine mutations, and more subtle effects for supporting residues like glutamate.
Substrate specificity comparisons between P. putida argC1 and other AGPRs require:
Enzyme preparation: Express and purify recombinant AGPRs from various organisms (e.g., E. coli, M. tuberculosis, plant sources) using identical tags and purification protocols.
Substrate panel testing: Assay each enzyme with a panel of substrates including:
Native substrate (N-acetyl-gamma-glutamyl-phosphate)
Structural analogs with modifications to the acetyl group
Analogs with altered phosphate positioning
Analogs with modified glutamyl backbones
Kinetic parameter determination: Measure Km, kcat, and kcat/Km ratios for each substrate with each enzyme.
Structural correlation: Analyze active site architectures through homology modeling or crystallography to identify residues responsible for specificity differences.
A typical data presentation might include:
| Enzyme Source | Substrate | Km (μM) | kcat (s^-1) | kcat/Km (M^-1s^-1) |
|---|---|---|---|---|
| P. putida | N-acetyl-γ-glutamyl-P | 100 | 10 | 1×10^5 |
| P. putida | N-propionyl-γ-glutamyl-P | 500 | 5 | 1×10^4 |
| E. coli | N-acetyl-γ-glutamyl-P | 150 | 12 | 8×10^4 |
| E. coli | N-propionyl-γ-glutamyl-P | 700 | 4 | 5.7×10^3 |
| M. tuberculosis | N-acetyl-γ-glutamyl-P | 80 | 8 | 1×10^5 |
| M. tuberculosis | N-propionyl-γ-glutamyl-P | 400 | 3 | 7.5×10^3 |
Such comparative analysis can reveal evolutionary adaptations in different organisms and suggest residues for targeted engineering to alter substrate specificity.
Advanced computational approaches for modeling argC1 catalytic mechanisms include:
Quantum mechanics/molecular mechanics (QM/MM) simulations: Apply quantum mechanical calculations to the active site region while treating the rest of the protein with molecular mechanics. This approach can accurately model bond breaking/forming events during catalysis, particularly the nucleophilic attack by the catalytic cysteine and subsequent proton transfer events .
Molecular dynamics simulations with enhanced sampling: Techniques such as umbrella sampling, metadynamics, or replica exchange can help overcome energy barriers to observe rare events in catalysis.
Reaction coordinate mapping: Calculate the potential energy surface along proposed reaction coordinates to identify transition states and intermediates.
pKa calculations: Predict the protonation states of key catalytic residues (His, Cys, Glu) under varying pH conditions to understand the pH dependence of activity.
Docking and molecular dynamics of enzyme-substrate complexes: Model substrate binding orientations and dynamics prior to catalysis. This approach has successfully identified substrate interactions with key residues (His217, His219, Arg114) in MtbAGPR .
Implementation should include:
Multiple starting structures to ensure adequate sampling
Validation against experimental kinetic and structural data
Sensitivity analyses for key parameters
Consideration of protein dynamics, particularly the conformational change observed upon NADP+ binding
A robust high-throughput screening (HTS) protocol for argC1 inhibitors would include:
Assay development:
Primary assay: NADPH consumption measured by fluorescence (excitation: 340 nm, emission: 460 nm)
Counter-screen: Rule out compounds that interfere with NADPH fluorescence
Secondary assay: Orthogonal method such as LC-MS to detect product formation
Assay optimization:
Miniaturization to 384- or 1536-well format
Determination of optimal enzyme and substrate concentrations
DMSO tolerance testing (typically up to 1-2%)
Statistical validation (Z' factor >0.7 for robustness)
Screening workflow:
Primary screen at single concentration (10-50 μM)
Dose-response curves for hits (IC50 determination)
Mechanism of inhibition studies for confirmed hits
Selectivity profiling against related enzymes
Structural studies of enzyme-inhibitor complexes:
Co-crystallization or soaking experiments
Molecular modeling of binding modes
This approach can identify novel inhibitors that may have potential as antimicrobial agents, particularly against organisms that rely on the arginine biosynthetic pathway .
Engineering argC1 for enhanced catalytic efficiency requires a multi-faceted approach:
Structure-guided mutagenesis:
Target residues near but not directly in the active site
Focus on second-shell residues that influence positioning of catalytic groups
Examine loop regions that might affect substrate access or product release
Directed evolution strategies:
Error-prone PCR to generate random mutations
DNA shuffling with homologous AGPRs from other organisms
Selection based on complementation of arginine auxotrophy
High-throughput screening for NADPH consumption
Semi-rational approaches:
Consensus sequence analysis across diverse AGPRs
Ancestral sequence reconstruction
Computational design of stabilizing mutations
Multi-parameter optimization:
Balance activity improvements with protein stability
Consider enzyme expression levels and solubility
Optimize for desired reaction conditions (pH, temperature)
Successful engineering might achieve 5-20 fold improvements in kcat/Km while maintaining or improving protein stability. Researchers should characterize engineered variants through detailed kinetic analysis, thermal stability measurements, and structural studies to understand the molecular basis of improved performance.
To investigate argC1's role in metabolic flux and regulation:
Gene knockout and complementation studies:
Generate argC1 deletion mutants
Complement with wild-type or mutant variants under different promoters
Assess growth phenotypes under varying nutrient conditions
Metabolic flux analysis:
Use 13C-labeled glutamate or glucose
Measure isotope distribution in pathway intermediates and products
Apply metabolic flux models to quantify pathway activity
Transcriptional regulation analysis:
RNA-seq to monitor gene expression changes in response to arginine availability
ChIP-seq to identify transcription factor binding sites
Reporter gene assays to quantify promoter activity
Protein-level regulation:
Measure enzyme levels using targeted proteomics
Investigate post-translational modifications
Assess protein-protein interactions that might modulate activity
In vivo enzyme activity:
Develop methods to measure argC1 activity in cell extracts
Compare in vitro and in vivo activities under different conditions
Use metabolite concentration data to estimate in vivo flux control coefficients