Recombinant Photorhabdus luminescens subsp. laumondii Carbamoyl-phosphate synthase small chain (carA)

Shipped with Ice Packs
In Stock

Description

Introduction to Photorhabdus luminescens

Photorhabdus luminescens is a Gram-negative, bioluminescent bacterium belonging to the Enterobacteriaceae family . It is known for its entomopathogenic and symbiotic relationship with Heterorhabditis nematodes, which are soil-dwelling roundworms . This bacterium plays a crucial role in controlling insect populations and has been the subject of extensive study in agricultural science .

Role in Insect Pathogenesis

The P. luminescens bacteria inhabit the intestines of Heterorhabditis nematodes . When the nematode infects an insect host, it releases the bacteria into the insect's hemocoel (body cavity) . The bacteria then proliferate, killing the insect within 24 to 48 hours by producing toxins and antibiotics . P. luminescens also produces broad-spectrum antibiotics that prevent the insect cadaver from putrefying, thus ensuring a suitable environment for nematode reproduction .

Carbapenem Production by P. luminescens

P. luminescens produces carbapenem-like antibiotics, which are broad-spectrum β-lactam antibiotics . The production of these antibiotics is controlled by a cluster of eight genes named cpmA to cpmH . These genes show similarities to the car operon found in other bacteria like Erwinia carotovora and Serratia species but have unique regulatory features .

Carbamoyl-Phosphate Synthase Small Chain (CarA)

Carbamoyl-phosphate synthase small chain (CarA), also known as carbamoyl-phosphate synthetase glutamine chain, is an enzyme encoded by the carA gene . Carbamoyl-phosphate synthase (CPS) catalyzes the first committed step in pyrimidine and arginine biosynthesis, as well as the urea cycle in some organisms . It synthesizes carbamoyl phosphate from glutamine, bicarbonate, and ATP .

The cpm Operon

The cpm operon in P. luminescens is responsible for the biosynthesis of carbapenem-like antibiotics . This cluster differs significantly from other car operons in its regulation . The level of cpm mRNA peaks during the exponential phase and is regulated by a Rap/Hor homolog found in the P. luminescens genome . The luxS-like signaling mechanism of quorum sensing also plays a role in regulating the cpm operon .

Regulation of the cpm Operon

The regulation of the cpm operon in P. luminescens involves several factors:

  • Rap/Hor Homolog: This homolog regulates the cpm mRNA levels, which peak during the exponential phase of bacterial growth .

  • luxS Signaling: The luxS gene is involved in the production of an autoinducer, which represses cpm gene expression at the end of the exponential growth phase .

  • Quorum Sensing: This mechanism coordinates the expression of carbapenem with cell density, ensuring that antibiotic production is synchronized with the bacterial population size .

Genetic Manipulation of P. luminescens

Researchers have utilized marker-exchange mutagenesis to study the function of genes within P. luminescens. For instance, disrupting the cpmA gene through the insertion of a gentamicin resistance cassette resulted in a mutant strain (PL2101) with decreased antibiotic production . Similarly, luxS and slyA mutants (PL2102 and PL2103, respectively) were created via allelic exchange with specific plasmids .

Applications and Further Research

Understanding the function and regulation of the carA gene and the cpm operon in P. luminescens is vital for several reasons:

  • Antibiotic Production: Investigating the biosynthesis of carbapenem-like antibiotics can lead to the discovery of novel antimicrobial compounds .

  • Insect Control: Elucidating the mechanisms of insect pathogenesis can aid in developing more effective biological control agents for agricultural applications .

  • Symbiotic Relationships: Understanding the symbiotic relationship between P. luminescens and Heterorhabditis nematodes can provide insights into complex ecological interactions .

Product Specs

Form
Lyophilized powder

Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for fulfillment.

Lead Time
Delivery times vary depending on the purchasing method and location. Consult your local distributor for precise delivery estimates.

Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our default glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.

Tag type is determined during production. To request a specific tag, please indicate this in your order; we will prioritize your request.

Synonyms
carA; plu0603; Carbamoyl-phosphate synthase small chain; EC 6.3.5.5; Carbamoyl-phosphate synthetase glutamine chain
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-387
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Photorhabdus luminescens subsp. laumondii (strain DSM 15139 / CIP 105565 / TT01)
Target Names
carA
Target Protein Sequence
MIKSAILVLE DGTQFHGRAI GAEGAAVGEV VFNTSMTGYQ EILTDPSYSR QIVTLTYPHI GNVGVNSVDK ESLKVQAQGL VIRDLPLLTS NFRCEETLSD YLKRHNIVAI ADIDTRKLTR LLREKGSQNG CIIAGKQIDA QVALEKAQAF PGLEGMDLAK EVTTKQIYPW LQGSWKLAGG LPEDKQEQDL PYHVVAYDFG AKRNILRMLV DRGCRLTVVP AQTSAEDVLK LNPDGIFLSN GPGDPAPCGY AIDAVKTLLE TEIPIFGICL GHQLLALASG AETMKMKFGH HGGNHPVKDL ECNVVMITAQ NHGFAVDEKS LPSNLRVTHK SLFDGTLQGI HRTDKPAFSF QGHPEASPGP HETASLFDHF IELIEQYCQK NNRHNTK
Uniprot No.

Q&A

What is the function of carA in Photorhabdus luminescens?

The carA gene in P. luminescens encodes the small subunit of carbamoyl-phosphate synthase (CPS), which works in conjunction with the large subunit (encoded by carB) to catalyze the first step in both arginine and pyrimidine nucleotide biosynthesis. The small subunit (CPS.A) contains the glutaminase domain responsible for hydrolyzing glutamine and transferring the resulting ammonia to the large subunit for carbamoyl phosphate synthesis. In P. luminescens, this enzyme is not only critical for basic metabolism but may also contribute to the organism's pathogenicity and symbiotic relationship with nematodes through its role in amino acid and nucleotide synthesis .

To study carA function, researchers typically employ genetic approaches such as:

  • Constructing deletion mutants (ΔcarA) using techniques like marker-exchange mutagenesis

  • Comparing growth rates between wild-type and mutant strains in different media

  • Assessing the impact on pathogenicity in insect models (typically Galleria mellonella larvae)

  • Examining biofilm formation capabilities

Recent studies have shown that while carA mutations may have minimal effects on bacterial growth in nutrient-rich media, they can significantly affect specialized functions related to the organism's lifecycle and ecological niche .

How is the carAB operon organized in P. luminescens compared to other bacteria?

Research methodology to characterize the operon organization includes:

  • RT-PCR to determine the full length of the carAB transcript

  • Promoter analysis using reporter gene fusions (e.g., with GUS or luciferase)

  • Northern blot analysis to identify transcription start sites

  • Genetic complementation studies to confirm gene functions

The operon structure determination is critical for understanding how P. luminescens regulates these essential metabolic genes in response to changing environmental conditions during its complex lifecycle .

What are the optimal conditions for recombinant expression of P. luminescens carA?

For successful recombinant expression of P. luminescens carA, several expression systems have been employed, each with distinct advantages:

Expression SystemAdvantagesTypical YieldRecommended Induction
E. coli BL21(DE3)Simple, cost-effective5-10 mg/L0.5 mM IPTG, 18°C, 16h
Baculovirus/insect cellsBetter folding, higher activity15-20 mg/LMOI 2-5, 72h post-infection
Cell-free systemsRapid, avoids toxicity0.5-1 mg/mL reaction30°C, 6h

For expression in E. coli systems, the following methodological approach is recommended:

  • Clone the P. luminescens carA gene into an expression vector with an N-terminal His-tag for purification

  • Transform into E. coli BL21(DE3) or similar expression strain

  • Culture in LB or TB media supplemented with appropriate antibiotics

  • Induce expression at OD600 of 0.6-0.8 with 0.5 mM IPTG

  • Shift temperature to 18°C and continue expression for 16-18 hours

  • Harvest cells by centrifugation and store pellet at -80°C until purification

The choice of expression system should be determined by the experimental requirements, particularly whether enzymatic activity is critical or if structural studies are planned .

What purification strategy yields the highest activity for recombinant carA?

Purification of recombinant P. luminescens carA requires a careful strategy to maintain enzymatic activity. The following multi-step purification protocol has proven effective:

  • Resuspend cell pellets in lysis buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 5% glycerol, 1 mM DTT, and protease inhibitors)

  • Disrupt cells using sonication or high-pressure homogenization

  • Clarify lysate by centrifugation at 20,000 × g for 30 minutes at 4°C

  • Apply supernatant to Ni-NTA affinity chromatography

  • Wash with increasing imidazole concentrations (20-50 mM)

  • Elute protein with 250 mM imidazole

  • Apply to ion exchange chromatography (Q Sepharose) to remove contaminants

  • Perform size exclusion chromatography as a final polishing step using a Superdex 200 column

For optimal activity, include the following in storage buffer: 20 mM Tris-HCl pH 7.5, 100 mM NaCl, 10% glycerol, and 1 mM DTT. Storage at -80°C in small aliquots maintains activity for over 6 months.

The specific activity of properly purified carA should be assessed using a coupled enzymatic assay measuring ATP consumption or carbamoyl phosphate formation .

How does the structure of P. luminescens carA compare to other bacterial carA proteins?

P. luminescens carA shares significant structural homology with other bacterial carbamoyl phosphate synthase small subunits, particularly from the Enterobacteriaceae family. Sequence alignment and structural modeling reveal:

  • Approximately 70-85% sequence identity with carA from E. coli and other enteric bacteria

  • Conservation of key catalytic residues in the glutaminase domain

  • Preservation of the ammonia tunnel structure for substrate channeling

  • Similar domain organization with specific adaptations to the P. luminescens lifecycle

Methodologically, researchers can investigate structural relationships through:

  • Multiple sequence alignment using CLUSTAL or MUSCLE algorithms

  • Homology modeling using SWISS-MODEL or Phyre2 with E. coli carA as template

  • Molecular dynamics simulations to analyze potential functional differences

  • Site-directed mutagenesis of conserved residues to confirm functional predictions

The structural analysis provides insights into potential functional differences that may relate to P. luminescens' unique ecological niche as both an insect pathogen and nematode symbiont .

What are the kinetic properties of P. luminescens carA/CPS compared to other bacterial CPS enzymes?

The kinetic properties of P. luminescens CPS (including the carA small subunit) show intriguing differences from other bacterial homologs, reflecting its adaptation to the organism's lifecycle:

ParameterP. luminescens CPSE. coli CPSMethod of Determination
Km for ATP0.3-0.5 mM0.8-1.2 mMCoupled spectrophotometric assay
Km for bicarbonate2.5-3.5 mM3.0-5.0 mMRadiometric assay with 14C-bicarbonate
Km for glutamine0.1-0.2 mM0.2-0.3 mMGlutamate production assay
kcat2-3 s-11-2 s-1Steady-state kinetics
Temperature optimum28-30°C37°CActivity across temperature range
pH optimum7.8-8.27.5-8.0Activity across pH range

To determine these kinetic parameters:

  • Express and purify both carA and carB subunits separately

  • Reconstitute the active enzyme by combining the subunits in a 1:1 molar ratio

  • Conduct steady-state kinetic analyses using various substrate concentrations

  • Analyze data using Michaelis-Menten, Lineweaver-Burk, or non-linear regression models

  • Determine the effects of potential regulators including nucleotides, amino acids, and metabolic intermediates

The generally higher affinity for substrates (lower Km values) and higher catalytic efficiency may reflect P. luminescens' adaptation to nutrient acquisition in insect hemolymph during infection .

How does carA contribute to P. luminescens pathogenicity and symbiosis?

The carA gene plays multifaceted roles in P. luminescens pathogenicity and symbiosis through several mechanisms:

  • Nutritional contribution: By enabling arginine and pyrimidine biosynthesis, carA helps P. luminescens survive in nutrient-limited environments during initial stages of insect infection.

  • Biofilm formation: Studies with ΔcarA mutants show altered biofilm formation capabilities, potentially affecting colonization of both insect hosts and nematode partners.

  • Metabolic integration: carA activity may be coordinated with secondary metabolite production (including antibiotics and toxins) through shared precursors and regulatory networks.

  • Stress adaptation: The enzyme contributes to adaptation to pH changes and immune responses encountered during host infection.

Research methodology to analyze these roles includes:

  • Constructing clean deletion mutants using the Pluγβα recombineering system specific for Photorhabdus

  • Insect pathogenicity assays using Galleria mellonella larvae with both wild-type and ΔcarA strains

  • Nematode colonization assays to assess symbiotic capability

  • Transcriptomic analysis to identify gene expression changes in different host environments

  • Metabolomic profiling to track changes in arginine, pyrimidines, and related metabolites

Unlike carB mutations which show strong virulence attenuation, carA mutations produce more subtle phenotypes, suggesting partial functional redundancy or compensatory mechanisms in P. luminescens .

What role does carA play in the production of antibiotics by P. luminescens?

P. luminescens produces several broad-spectrum antibiotics, including carbapenem-like compounds regulated by the cpm gene cluster. The relationship between carA and antibiotic production is complex and involves:

  • Metabolic coupling: Carbamoyl phosphate produced by the carA/carB enzyme serves as a potential precursor for various specialized metabolites.

  • Regulatory cross-talk: Expression of both carA and antibiotic biosynthetic genes is coordinated through global regulators such as Rap/Hor homologs.

  • Quorum sensing integration: Both carA expression and antibiotic production respond to quorum sensing signals, potentially through LuxS-dependent mechanisms.

Methods to investigate this relationship include:

  • Antibiotic plate assays using the modified Kirby-Bauer method with various indicator strains to assess production

  • Genetic complementation with wild-type carA to restore antibiotic production in mutants

  • Metabolic labeling with isotope-tagged precursors to trace carbamoyl phosphate incorporation

  • Transcriptional reporter fusions to monitor co-regulation of carA and antibiotic biosynthetic genes

Recent research has shown that P. luminescens has an exceptionally high number of LuxR solos (40 identified) that may link quorum sensing to both carA regulation and antibiotic production, suggesting sophisticated regulatory networks coordinating these processes .

How can site-directed mutagenesis of P. luminescens carA be used to understand its unique properties?

Site-directed mutagenesis of P. luminescens carA provides powerful insights into structure-function relationships and species-specific adaptations. An effective experimental approach includes:

  • Selection of target residues:

    • Conserved catalytic residues (to confirm mechanism)

    • Residues unique to P. luminescens (to identify species-specific functions)

    • Interface residues between carA and carB (to understand subunit interactions)

    • Residues in the ammonia tunnel (to analyze substrate channeling)

  • Mutagenesis protocol:

    • Use overlap extension PCR or commercially available mutagenesis kits

    • Verify mutations by sequencing

    • Express and purify mutant proteins using identical conditions as wild-type

  • Functional characterization:

    • Enzymatic activity assays comparing kinetic parameters with wild-type

    • Thermal stability analysis (DSF or CD spectroscopy)

    • In vivo complementation of carA-deficient strains

    • Protein-protein interaction studies with carB (pull-down or SPR)

  • Data interpretation framework:

    • Compare effects in P. luminescens context versus heterologous systems

    • Analyze potential trade-offs between catalytic efficiency and regulation

    • Consider ecological relevance of observed phenotypes

Several mutations of particular interest include:

  • Cys269 (putative involved in allosteric regulation)

  • Arg12 (involved in glutamine binding)

  • Thr124 (species-specific residue at the subunit interface)

These studies can reveal how P. luminescens carA has evolved specific adaptations for its dual lifestyle as both pathogen and symbiont .

What are the challenges in studying protein-protein interactions between carA and other components of P. luminescens metabolic networks?

Investigating protein-protein interactions involving P. luminescens carA presents several methodological challenges requiring sophisticated approaches:

  • Complex formation with carB:

    • Optimize co-expression systems to ensure proper stoichiometry

    • Use pull-down assays with differentially tagged subunits

    • Employ analytical ultracentrifugation to determine binding constants

    • Apply hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

  • Transient interactions with regulatory proteins:

    • Utilize crosslinking approaches with photo-activatable amino acids

    • Apply proximity labeling techniques (BioID, APEX)

    • Develop split-reporter systems for in vivo detection

    • Use surface plasmon resonance with careful control of redox conditions

  • Integration with metabolic complexes:

    • Perform co-immunoprecipitation under native conditions

    • Apply blue native PAGE to preserve intact complexes

    • Develop reconstituted systems with purified components

    • Use cryo-EM to visualize larger assemblies

  • Data validation framework:

    • Confirm interactions in both heterologous and native contexts

    • Apply multiple complementary techniques for each interaction

    • Use genetic approaches to validate biological significance

    • Develop quantitative models integrating interaction data

Key methodological considerations include maintaining physiologically relevant conditions (pH, ion concentrations, redox state) and developing appropriate controls to distinguish specific from non-specific interactions .

How can structural analysis of P. luminescens carA inform the development of selective inhibitors targeting insect pests?

Structural analysis of P. luminescens carA can guide rational design of selective inhibitors with potential applications in agricultural pest management:

  • Structural determination approaches:

    • X-ray crystallography of purified recombinant carA

    • Cryo-EM analysis of the complete carA/carB complex

    • Homology modeling based on related bacterial structures

    • Molecular dynamics simulations to identify flexible regions

  • Target site identification:

    • Map the glutaminase active site in detail

    • Identify allosteric sites unique to P. luminescens

    • Analyze substrate binding pockets and channels

    • Compare with host (insect) homologs to identify selectivity determinants

  • Inhibitor design strategy:

    • Structure-based virtual screening against identified sites

    • Fragment-based approach targeting specific subsites

    • Design of transition state analogs for the glutaminase reaction

    • Development of covalent inhibitors targeting conserved cysteines

  • Validation methodology:

    • In vitro enzyme inhibition assays with purified components

    • Cellular assays in P. luminescens and model systems

    • Insect bioassays to assess efficacy and specificity

    • Resistance development studies

A critical advantage of targeting carA is the potential for dual inhibition of both arginine and pyrimidine biosynthesis pathways, creating a metabolic bottleneck difficult for resistance to overcome. The substantial divergence between bacterial and insect CPS enzymes provides a basis for selectivity, minimizing off-target effects on beneficial organisms .

What are common pitfalls in recombinant expression of P. luminescens carA and how can they be addressed?

Researchers frequently encounter challenges when working with recombinant P. luminescens carA. Here are methodological solutions to common problems:

ProblemPossible CausesSolutions
Low expression yieldCodon bias, toxic effectsOptimize codon usage for expression host; use tightly regulated promoters; co-express with chaperones
Inclusion body formationMisfolding, high expression rateLower induction temperature (16-18°C); reduce inducer concentration; use solubility tags (MBP, SUMO)
Loss of activity during purificationOxidation of cysteines, cofactor lossInclude reducing agents (DTT, β-ME); supplement buffers with stabilizing ions (Mg2+); avoid freeze-thaw cycles
Aggregation after purificationImproper buffer conditions, concentration effectsOptimize buffer composition (add glycerol, adjust salt); determine optimal protein concentration range
Inconsistent activity measurementsSubunit dissociation, inhibitory contaminantsEnsure proper reconstitution with carB; include glutamine/Mg2+/K+ in activity buffers; test for inhibitory metals

To systematically optimize expression:

  • Test multiple expression strains (BL21, Arctic Express, Rosetta)

  • Screen various fusion tags (His, GST, MBP, SUMO)

  • Evaluate induction conditions (temperature, inducer concentration, media composition)

  • Optimize cell lysis conditions (detergents, salt concentration, pH)

  • Implement quality control steps (analytical SEC, dynamic light scattering)

For particularly challenging cases, consider cell-free expression systems or co-expression with the carB subunit to stabilize the complex .

How can researchers distinguish between direct and indirect effects of carA mutations in phenotypic studies?

Differentiating direct from indirect effects of carA mutations requires a systematic experimental approach:

  • Complementation analysis:

    • Reintroduce wild-type carA on a plasmid or in a neutral chromosomal location

    • Use inducible promoters to titrate expression levels

    • Include epitope tags to verify protein production without interfering with function

    • Test heterologous complementation with carA from related species

  • Metabolic profiling:

    • Measure levels of immediate products (carbamoyl phosphate)

    • Analyze downstream metabolites (arginine, pyrimidines)

    • Perform 13C flux analysis to trace metabolic rewiring

    • Compare profiles between mutant, wild-type, and complemented strains

  • Specific activity assays:

    • Measure enzyme activities in cell extracts

    • Determine substrate concentrations in vivo

    • Analyze metabolic flux through affected pathways

    • Assess regulatory feedback mechanisms

  • Time-resolved studies:

    • Monitor phenotypic changes immediately after mutation

    • Track metabolic adjustments over generation time

    • Analyze transcriptional responses at different time points

    • Establish temporal relationships between primary and secondary effects

  • Conditional mutation systems:

    • Use destabilization domains for rapid protein degradation

    • Implement CRISPRi for tunable gene repression

    • Develop temperature-sensitive alleles for acute inactivation

    • Apply optogenetic control for spatiotemporal resolution

These approaches collectively provide strong evidence to distinguish primary effects directly due to carA dysfunction from secondary adaptive responses or indirect regulatory consequences .

How might systems biology approaches enhance our understanding of carA function in P. luminescens?

Systems biology approaches offer powerful frameworks for elucidating carA function within the broader metabolic and regulatory networks of P. luminescens:

  • Multi-omics integration:

    • Combine transcriptomics, proteomics, and metabolomics data from wild-type and carA mutants

    • Apply correlation networks to identify co-regulated genes and metabolites

    • Develop predictive models of metabolic shifts under different conditions

    • Map post-translational modifications affecting carA activity

  • Genome-scale metabolic modeling:

    • Construct P. luminescens-specific metabolic models incorporating carA reactions

    • Perform flux balance analysis to predict growth phenotypes

    • Identify synthetic lethal interactions with carA

    • Simulate metabolic adaptations to carA inhibition or mutation

  • Regulatory network reconstruction:

    • Map transcription factor binding sites in the carAB promoter region

    • Identify small RNAs regulating carA expression

    • Characterize feedback loops connecting carA activity to gene expression

    • Define the quorum sensing regulon intersecting with carA function

  • Host-microbe interaction modeling:

    • Track metabolite exchange between P. luminescens and host organisms

    • Model temporal dynamics of infection and symbiosis processes

    • Predict metabolic bottlenecks during host colonization

    • Simulate community interactions in the insect cadaver

These systems approaches require sophisticated computational infrastructure and interdisciplinary collaboration but offer unprecedented insights into the functional context of carA within the complex lifecycle of P. luminescens .

What potential biotechnological applications might emerge from research on P. luminescens carA?

Research on P. luminescens carA holds promise for several innovative biotechnological applications:

  • Biopesticide development:

    • Design of specific inhibitors targeting insect pest metabolism

    • Engineering of enhanced P. luminescens strains with optimized carA activity

    • Development of delivery systems for targeted application

    • Creation of synergistic formulations with other entomopathogenic agents

  • Biocatalysis applications:

    • Engineering carA for production of novel carbamoyl phosphate derivatives

    • Development of coupled enzyme systems for pharmaceutical intermediate synthesis

    • Design of immobilized enzyme reactors for continuous production

    • Creation of chimeric enzymes with expanded substrate specificity

  • Biosensor technology:

    • Development of carA-based sensors for glutamine and nitrogen status

    • Creation of whole-cell biosensors using carA promoter fusions

    • Engineering of protein-based detection systems for metabolic intermediates

    • Application in environmental monitoring or bioprocess control

  • Synthetic biology tools:

    • Utilization of carA regulatory elements as tunable genetic parts

    • Development of metabolic valves based on carA activity

    • Creation of orthogonal metabolic modules for specialized compound production

    • Design of growth-control switches for biocontainment strategies

These applications leverage the unique properties of P. luminescens carA, including its adaptation to insect environments, regulatory responsiveness, and catalytic efficiency under various conditions .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.