Recombinant Nitrosomonas europaea Bifunctional protein GlmU (glmU)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Nitrosomonas europaea Bifunctional Protein GlmU (glmU)

The Recombinant Nitrosomonas europaea Bifunctional Protein GlmU (glmU) is a recombinant protein derived from the bacterium Nitrosomonas europaea. This protein is crucial for various biochemical processes within the cell, particularly in the synthesis and modification of peptidoglycan, a key component of bacterial cell walls. The glmU gene encodes a bifunctional enzyme that includes two domains: UDP-N-acetylglucosamine pyrophosphorylase and N-acetylglucosamine-1-phosphate uridyltransferase. These enzymatic activities are essential for the synthesis of UDP-N-acetylglucosamine, a precursor molecule in peptidoglycan biosynthesis.

Characteristics and Function of GlmU

  • Enzymatic Activities: The GlmU protein exhibits two main enzymatic activities:

    • UDP-N-acetylglucosamine Pyrophosphorylase: This activity involves the synthesis of UDP-N-acetylglucosamine from N-acetylglucosamine-1-phosphate and UTP.

    • N-acetylglucosamine-1-phosphate Uridyltransferase: This activity catalyzes the conversion of N-acetylglucosamine-1-phosphate to UDP-N-acetylglucosamine.

  • Sequence and Structure: The sequence of the GlmU protein from Nitrosomonas europaea is well-documented, with a specific amino acid sequence that defines its structure and function .

Data Table: Characteristics of Recombinant GlmU Protein

CharacteristicDescription
Protein TypeRecombinant Protein
Immunogen SpeciesNitrosomonas europaea
Purity>85% (SDS-PAGE)
Uniprot No.Q82XP7
Expression RegionFull-length protein (1-458 amino acids)
Storage ConditionsLiquid: 6 months at -20°C/-80°C; Lyophilized: 12 months at -20°C/-80°C

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order remarks if necessary. We will accommodate your request whenever possible.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
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.
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 consolidate 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 standard glycerol concentration is 50% and may serve as a guideline for your preparations.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and inherent protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms 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
The tag type will be determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
glmU; NE0208Bifunctional protein GlmU [Includes: UDP-N-acetylglucosamine pyrophosphorylase; EC 2.7.7.23; N-acetylglucosamine-1-phosphate uridyltransferase); Glucosamine-1-phosphate N-acetyltransferase; EC 2.3.1.157)]
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-458
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nitrosomonas europaea (strain ATCC 19718 / CIP 103999 / KCTC 2705 / NBRC 14298)
Target Names
glmU
Target Protein Sequence
MLQVDVVILA AGMGKRMCSS LPKVLHPLAG KPILSHVLDI ARTLSPERIC VVFGYGGELV RQVIGDHSDL IWVKQAQQLG TGHAVKQALP YLGNKGVTLV LFGDVPLVKS DTLKALIEKA REDNLVLLTV ELDNPTGYGR IVRDPVTNRI QAIVEEKDAS QSQKKIREIN TGIMVLPNGR LGNWLDNLSD ANTQGEYYLT DIIAMAVDAG IPIETSSPAS DWEVSGVNDK IQLSILERAH QQDTANRLME QGVMFADPAR FDVRGRLVCG NDVEIDINCI FEGNVRLGNN VKIHANCILR NVIVSDGSVV HPFSLIEDAE VGKNCRIGPY ARIRPGTQLD DAVHVGNFVE IKNSHIASES KVNHLSYVGD TEMGRRVNIG AGAITCNYDG AFKHRTVIED DVFIGSDTQL VAPVTVARGS TIGAGSTITR DTPEGQLTLS RTKQTSIANW KRPRKDRN
Uniprot No.

Target Background

Function
This bifunctional GlmU protein from *Nitrosomonas europaea* catalyzes the final two steps in the de novo UDP-N-acetylglucosamine (UDP-GlcNAc) biosynthetic pathway. The C-terminal domain acetylates glucosamine-1-phosphate (GlcN-1-P) using acetyl-CoA to produce N-acetylglucosamine-1-phosphate (GlcNAc-1-P). Subsequently, the N-terminal domain catalyzes the transfer of uridine 5-monophosphate (from UTP) to GlcNAc-1-P, yielding UDP-GlcNAc.
Database Links

KEGG: neu:NE0208

STRING: 228410.NE0208

Protein Families
N-acetylglucosamine-1-phosphate uridyltransferase family; Transferase hexapeptide repeat family
Subcellular Location
Cytoplasm.

Q&A

What is the bifunctional protein GlmU in Nitrosomonas europaea and what are its primary functions?

GlmU (Bifunctional protein GlmU) in Nitrosomonas europaea is a cytoplasmic enzyme that catalyzes the last two sequential reactions in the de novo biosynthetic pathway for UDP-N-acetylglucosamine (UDP-GlcNAc) . This bifunctional enzyme possesses two distinct catalytic activities:

  • Acetyltransferase activity (C-terminal domain): Catalyzes the transfer of an acetyl group from acetyl coenzyme A to glucosamine-1-phosphate (GlcN-1-P), producing N-acetylglucosamine-1-phosphate (GlcNAc-1-P) .

  • Uridyltransferase activity (N-terminal domain): Catalyzes the conversion of GlcNAc-1-P into UDP-GlcNAc by transferring uridine 5-monophosphate from uridine 5-triphosphate .

The product UDP-GlcNAc serves as an essential precursor for biosynthetic pathways of peptidoglycan and other bacterial cell wall components, making GlmU crucial for bacterial cell survival and integrity .

What is the structural organization of GlmU and how does it relate to its bifunctional nature?

The GlmU protein exhibits a distinct two-domain architecture connected by a long α-helical arm, directly corresponding to its bifunctional nature . The crystal structure analysis reveals:

  • N-terminal domain: Resembles the dinucleotide-binding Rossmann fold, responsible for the uridyltransferase activity .

  • C-terminal domain: Adopts a left-handed parallel β-helix structure (LβH) as found in homologous bacterial acetyltransferases, responsible for the acetyltransferase activity .

  • Connecting region: A long α-helical arm that links the two functional domains .

The quaternary structure shows that three GlmU molecules assemble into a trimeric arrangement with tightly packed parallel LβH domains. The long α-helical linkers are seated on top of this arrangement while the N-terminal domains project away from the 3-fold axis . This structural organization allows each domain to function independently while being part of the same polypeptide chain.

Why is N. europaea an important model organism for studying bacterial cell wall biosynthesis enzymes?

Nitrosomonas europaea serves as an excellent model organism for studying bacterial cell wall biosynthesis enzymes for several scientific reasons:

  • Well-defined metabolism: N. europaea has a well-characterized ammonia metabolism, making it easier to study specific pathways in isolation .

  • Established genetic tools: A wide range of physiological and transcriptional tools are available for N. europaea, facilitating detailed investigation of gene function and regulation .

  • Complete genome sequence: As the first ammonia-oxidizing bacterium (AOB) to have its genome sequenced (strain ATCC 19718), it provides a comprehensive genetic background for research .

  • Environmental significance: N. europaea plays a crucial role in the nitrogen cycle, making research on its cell wall biosynthesis relevant to environmental microbiology and wastewater treatment .

  • Adaptability to various growth conditions: N. europaea can be cultured in batch, continuous, and biofilm conditions, allowing for versatile experimental setups .

These characteristics make N. europaea particularly valuable for studying enzymes like GlmU that are involved in essential cellular processes.

What expression systems have been optimized for recombinant production of N. europaea GlmU?

While the search results don't specifically detail optimized expression systems for recombinant N. europaea GlmU, we can draw on protocols established for expressing recombinant proteins in N. europaea and similar bacterial systems:

Table 1: Comparison of Expression Systems for Recombinant N. europaea Proteins

Expression SystemAdvantagesLimitationsNotable Success
N. europaea self-expressionNative folding and post-translational modificationsSlow growth rate, lower yieldGFP expression using native promoters (mbla, clpB)
E. coli expression systemsHigh yield, rapid growth, well-established protocolsPotential folding issues, lack of specific post-translational modificationsStructural studies of similar bacterial GlmU proteins
Cell-free protein synthesisRapid production, avoids toxicity issuesHigher cost, lower yieldNot specifically documented for N. europaea GlmU

For recombinant expression in N. europaea itself, researchers have successfully used vectors with inducible promoters as demonstrated by the construction of recombinant N. europaea expressing green fluorescent protein (GFP) . When designing expression systems for GlmU, careful consideration should be given to maintaining the proper folding of both domains to preserve bifunctional activity.

How can site-directed mutagenesis be used to investigate the catalytic mechanism of GlmU?

Site-directed mutagenesis is a powerful approach for investigating the catalytic mechanism of bifunctional enzymes like GlmU. Based on the crystal structure and functional analysis of GlmU, researchers can target specific amino acid residues suspected to be involved in substrate binding or catalysis .

Methodological approach for GlmU mutagenesis studies:

  • Target identification: Based on the crystal structure of GlmU complexed with UDP-GlcNAc, identify conserved residues in the active sites of both domains .

  • Mutagenesis design: Create point mutations that:

    • Alter residues in the N-terminal domain to specifically affect uridyltransferase activity

    • Modify residues in the C-terminal domain to affect acetyltransferase activity

    • Change residues in the connecting α-helical arm to investigate domain communication

  • Activity assays: Measure both enzymatic activities separately to determine the effect of mutations:

    • Acetyltransferase activity: Monitor the acetylation of glucosamine-1-phosphate

    • Uridyltransferase activity: Assess the conversion of GlcNAc-1-P to UDP-GlcNAc

  • Structural analysis: Perform crystallographic studies of mutant proteins to correlate functional changes with structural alterations

This approach has proven valuable for understanding catalytic mechanisms in homologous GlmU proteins and can be adapted specifically for N. europaea GlmU .

How does GlmU from N. europaea compare structurally and functionally with GlmU from other bacterial species?

Comparative analysis of GlmU from N. europaea with homologs from other bacterial species reveals important structural and functional conservation as well as species-specific adaptations.

Table 2: Comparative Analysis of GlmU Across Bacterial Species

FeatureN. europaea GlmUE. coli GlmUOther Bacterial GlmUSignificance
Protein length458 amino acids ~450 amino acidsVariable (430-470 aa)Core functional regions conserved across species
Domain organizationBifunctional: N-terminal uridyltransferase, C-terminal acetyltransferase Same bifunctional arrangementGenerally conserved bifunctional arrangementEvolutionary conservation of essential function
Quaternary structureTrimeric assembly TrimericPrimarily trimericConserved oligomeric state important for function
C-terminal domainLeft-handed parallel β-helix (LβH) LβH structureLβH structure in most speciesHighly conserved fold for acetyltransferase activity
Sequence conservationReferenceHigh similarity in catalytic residuesVariable conservation in non-catalytic regionsCatalytic mechanism preserved across diverse bacteria

What are the optimal conditions for expressing and purifying recombinant N. europaea GlmU?

Based on the available information and established protocols for similar proteins, the following methodology can be employed for optimal expression and purification of recombinant N. europaea GlmU:

Expression protocol:

  • Vector selection: Use of pET-based expression vectors with T7 promoter systems has proven effective for many bacterial enzymes .

  • Host strain: E. coli BL21(DE3) or its derivatives are recommended for high-level expression of recombinant proteins.

  • Induction conditions: Optimize IPTG concentration (typically 0.1-1.0 mM) and induction temperature (16-30°C). Lower temperatures (16-18°C) often improve the solubility of multi-domain proteins like GlmU.

  • Expression duration: Extended expression periods (16-20 hours) at lower temperatures can enhance proper folding of the bifunctional enzyme.

Purification strategy:

  • Initial lysis: Sonication or high-pressure homogenization in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, and protease inhibitors.

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using His-tagged GlmU.

  • Secondary purification: Ion exchange chromatography to separate fully active enzyme from partially folded species.

  • Final polishing: Size exclusion chromatography to isolate the properly assembled trimeric form of GlmU, which is crucial for full activity .

Storage conditions:
Buffer containing 25 mM Tris-HCl pH 7.5, 150 mM NaCl, and 10% glycerol at -80°C has been found effective for long-term storage of similar enzymes while maintaining bifunctional activity.

How can the two distinct enzymatic activities of GlmU be assayed separately?

The bifunctional nature of GlmU necessitates specific assay methods to evaluate each catalytic activity independently:

Acetyltransferase activity (C-terminal domain):

  • Direct acetylation monitoring:

    • Substrate: Glucosamine-1-phosphate (GlcN-1-P) and acetyl-CoA

    • Detection: DTNB (5,5'-dithiobis-(2-nitrobenzoic acid)) method to quantify free CoA production

    • Measurement: Spectrophotometric detection at 412 nm

  • Coupled enzyme assay:

    • Link CoA production to NADH oxidation via auxiliary enzymes

    • Monitor decrease in absorbance at 340 nm

Uridyltransferase activity (N-terminal domain):

  • Direct product quantification:

    • Substrate: N-acetylglucosamine-1-phosphate (GlcNAc-1-P) and UTP

    • Detection: HPLC or capillary electrophoresis to measure UDP-GlcNAc formation

    • Alternatives: Radioactive [α-32P]UTP can be used for higher sensitivity

  • Pyrophosphate release measurement:

    • Couple pyrophosphate release to enzymatic reactions that produce colorimetric or fluorescent outputs

    • Commercial kits available for pyrophosphate detection

Assay conditions optimization:

  • Buffer: 50 mM Tris-HCl pH 7.5-8.0

  • Salt: 10-100 mM NaCl or KCl

  • Metal ions: Mg2+ (1-5 mM) for uridyltransferase activity

  • Temperature: Typically 25-37°C, with N. europaea enzymes often showing activity at lower temperatures

These separate assays enable researchers to investigate the impact of mutations, inhibitors, or environmental conditions on each specific activity of the bifunctional GlmU enzyme.

How can transcriptomic and proteomic approaches be used to study GlmU regulation in N. europaea?

Investigating GlmU regulation in N. europaea requires integrated transcriptomic and proteomic approaches that can reveal regulatory mechanisms at multiple levels:

Transcriptomic approaches:

  • RNA-Seq analysis: Similar to techniques used for studying other N. europaea genes under various conditions, RNA-Seq can reveal glmU transcriptional regulation under different environmental stressors .

  • Promoter analysis: Transcriptional fusions with reporter genes (like GFP) can be used to monitor glmU promoter activity under various conditions, as demonstrated for other N. europaea genes (mbla, clpB) .

  • RT-qPCR validation: Quantitative PCR can precisely measure glmU transcript levels in response to specific conditions that might affect cell wall synthesis.

  • Transcriptional regulator identification:

    • ChIP-seq to identify proteins that bind to the glmU promoter region

    • Differential expression analysis to identify co-regulated genes in the same pathway

Proteomic approaches:

  • Mass spectrometry-based quantification: Targeted proteomics using multiple reaction monitoring (MRM) can quantify GlmU protein levels under different growth conditions.

  • Post-translational modification analysis: Phosphoproteomics and other PTM-specific analyses can reveal regulatory modifications of GlmU.

  • Protein-protein interaction studies:

    • Co-immunoprecipitation coupled with mass spectrometry

    • Bacterial two-hybrid systems to identify interacting partners

Integrated analysis workflow:

  • Culture N. europaea under conditions relevant to cell wall synthesis regulation (nutrient limitation, stress conditions, growth phase transitions)

  • Collect samples for parallel RNA extraction and protein purification

  • Perform transcriptomic analysis (RNA-Seq, RT-qPCR) and proteomic analysis (LC-MS/MS)

  • Correlate changes in glmU transcript levels with GlmU protein abundance

  • Identify potential regulatory mechanisms by examining promoter regions and protein modifications

This multi-omics approach has been successfully applied to study gene regulation in N. europaea under oxygen-limited conditions and could be adapted specifically for investigating glmU regulation .

How can recombinant N. europaea GlmU be utilized in developing novel antibacterial agents?

The essential role of GlmU in bacterial cell wall biosynthesis makes it an attractive target for antibacterial drug development. The crystal structure of GlmU provides a three-dimensional template for structure-based drug design . Research approaches include:

  • Structure-based inhibitor design:

    • Virtual screening against the active sites of both domains

    • Fragment-based drug discovery targeting binding pockets identified in crystal structures

    • Rational design of transition state analogs for each catalytic activity

  • High-throughput screening approaches:

    • Biochemical assays using purified recombinant GlmU to screen compound libraries

    • Cell-based assays using N. europaea or heterologous systems with modified permeability

  • Dual-action inhibitor development:

    • Design compounds that simultaneously inhibit both enzymatic activities

    • Compare with single-domain inhibitors to evaluate synergistic effects

  • Domain-specific targeting strategies:

    • N-terminal domain (uridyltransferase) inhibitors designed to compete with UTP binding

    • C-terminal domain (acetyltransferase) inhibitors targeting the acetyl-CoA binding site

    • Allosteric inhibitors disrupting domain communication or trimer formation

Table 3: Potential GlmU Inhibitor Types and Their Mechanisms

Inhibitor TypeTarget SiteMechanismAdvantages
Competitive nucleotide analogsN-terminal active siteCompete with UTP bindingSpecificity due to structural differences from human enzymes
Acetyl-CoA mimeticsC-terminal active siteBlock acetyl donor bindingLeverage unique bacterial acetyltransferase structure
Transition state analogsCatalytic sitesMimic reaction intermediatesHigh affinity and specificity
Allosteric inhibitorsDomain interfaces or oligomerization sitesDisrupt protein dynamics or assemblyNovel mode of action less prone to resistance

The crystal structure of GlmU-UDP-GlcNAc complex provides critical insights into the residues involved in substrate binding and catalysis, offering specific targets for inhibitor design .

What is the role of GlmU in N. europaea's response to environmental stressors?

While the search results don't directly address GlmU's specific role in stress response, we can analyze how this essential cell wall biosynthesis enzyme might be involved in N. europaea's adaptation to environmental stressors:

  • Oxygen limitation response:
    N. europaea undergoes significant transcriptomic changes under oxygen-limited conditions . As cell wall integrity is crucial during stress, GlmU activity likely plays a role in maintaining cellular structure during adaptation to hypoxic environments.

  • Stress-responsive regulation:
    Studies have shown that N. europaea upregulates specific genes (such as mbla and clpB) in response to stressors like chloroform and hydrogen peroxide . Similar regulation might occur for glmU when cell wall remodeling is needed during stress response.

  • Integration with cellular stress responses:
    The essential nature of UDP-GlcNAc for cell wall biosynthesis suggests that GlmU activity must be maintained even under stress conditions. This may involve post-translational modifications or regulated expression to ensure continued cell wall maintenance.

  • Methodological approach for investigation:
    Similar to the approach used for investigating other stress biomarkers in N. europaea , researchers can:

    • Monitor glmU transcript and protein levels under various stressors

    • Create reporter strains with GFP fusions to the glmU promoter to visualize regulation

    • Assess phenotypic effects of glmU overexpression or controlled downregulation during stress

  • Potential connection to nitrification inhibition:
    As N. europaea is studied as a model for nitrification inhibition , investigating how inhibitors affect GlmU activity could reveal connections between cell wall biosynthesis and nitrification capacity under stress conditions.

Understanding GlmU's role in stress response could provide insights into bacterial adaptation mechanisms and potentially reveal new approaches for controlling nitrification in environmental applications.

How can biosensor technology using N. europaea be adapted to study GlmU function and inhibition?

Building upon the successful development of biosensors using recombinant N. europaea expressing green fluorescent protein (GFP) , similar approaches can be adapted to study GlmU function and inhibition:

Methodology for biosensor construction:

  • Identify promoters regulated by cell wall stress or UDP-GlcNAc availability

  • Create transcriptional fusions with reporter genes (gfp variants)

  • Transform N. europaea using established protocols similar to those used for pPRO/mbla4 and pPRO/clpb7 constructs

  • Validate biosensor response using known conditions that affect cell wall biosynthesis

Table 4: Comparison of Biosensor Designs for GlmU Studies

Biosensor TypeReporter SystemDetection MethodApplication
Transcriptional fusionglmU promoter-gfpFluorescence microscopy/spectroscopyMonitor natural regulation of glmU expression
Metabolite-responsiveSynthetic UDP-GlcNAc-sensing element with reporterFluorescence intensityMeasure in vivo GlmU activity
Dual-domain reporterSeparate reporters for each enzymatic activityRatiometric fluorescenceDistinguish inhibition mechanisms
Stress-responsiveCell wall stress promoters (e.g., mbla) with reporterConcentration-dependent fluorescenceScreen for compounds affecting cell wall integrity

These biosensor approaches would extend the proven technology of GFP expression in N. europaea to create valuable tools for studying GlmU biology and for screening potential inhibitors in a cellular context.

What emerging technologies could advance our understanding of GlmU structure-function relationships?

Several cutting-edge technologies offer promising approaches to deepen our understanding of GlmU structure-function relationships:

  • Cryo-electron microscopy (Cryo-EM):

    • Enables visualization of GlmU in different conformational states without crystallization

    • Can reveal dynamic aspects of domain interactions during catalytic cycles

    • Particularly valuable for capturing transient intermediate states

  • Time-resolved X-ray crystallography:

    • Captures structural snapshots during enzymatic reactions

    • Helps identify conformational changes during substrate binding and product release

    • Provides insights into the coordination between the two catalytic domains

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Maps protein dynamics and solvent accessibility changes during catalysis

    • Identifies regions involved in allosteric communication between domains

    • Complements static structural data with information about protein flexibility

  • Single-molecule FRET studies:

    • Monitors real-time conformational changes of individual GlmU molecules

    • Reveals potential heterogeneity in the population of enzyme molecules

    • Provides insights into the coordination between domains during catalysis

  • Integrative structural biology approaches:

    • Combines multiple experimental techniques (X-ray, NMR, SAXS, Cryo-EM) with computational modeling

    • Creates comprehensive structural models of GlmU in different functional states

    • Particularly valuable for understanding the trimeric assembly dynamics

These advanced technologies, when applied to N. europaea GlmU, could reveal crucial insights into how this bifunctional enzyme coordinates its two catalytic activities and how it might be effectively targeted for antimicrobial development .

How might CRISPR-Cas9 technology be utilized to study GlmU function in N. europaea?

CRISPR-Cas9 technology offers powerful approaches for investigating GlmU function in N. europaea, despite the challenges of genetic manipulation in this organism:

  • Precise genetic modifications:

    • Create point mutations in catalytic residues to separate the two enzymatic functions

    • Generate domain deletions to study the independence of each catalytic activity

    • Introduce tagged versions of GlmU for in vivo localization and interaction studies

  • Conditional expression systems:

    • Develop CRISPR interference (CRISPRi) systems to achieve tunable repression of glmU

    • Create inducible promoter replacements to control GlmU expression levels

    • Engineer synthetic regulatory circuits to study GlmU regulation in detail

  • Genome-wide interaction screens:

    • Perform CRISPR-based screens to identify genetic interactions with glmU

    • Discover compensatory pathways that activate when GlmU function is compromised

    • Map the genetic network connected to cell wall biosynthesis in N. europaea

  • In vivo biosensor integration:

    • Use CRISPR-mediated homologous recombination to integrate reporter constructs

    • Create knock-in reporters that maintain native regulation while enabling monitoring

    • Develop multiplexed reporter systems to study GlmU in conjunction with other stress response elements

Methodological considerations for CRISPR application in N. europaea:

  • Optimize transformation protocols specific to N. europaea

  • Develop specialized vectors for this ammonia-oxidizing bacterium

  • Consider the limited genetic tools currently available and adapt CRISPR systems accordingly

The implementation of CRISPR-Cas9 technology would significantly advance our ability to study GlmU function in its native context and could revolutionize genetic manipulation capabilities in N. europaea, similar to advances made in other bacterial systems.

What are the most significant unanswered questions about N. europaea GlmU that warrant further investigation?

Despite the considerable knowledge about bacterial GlmU enzymes in general, several significant questions about N. europaea GlmU remain unanswered and merit focused research:

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 2024 Thebiotek. All Rights Reserved.