Recombinant Nitrosomonas europaea Ribonuclease H (rnhA)

Shipped with Ice Packs
In Stock

Description

Overview of Ribonuclease H

Ribonuclease H (RNase H) enzymes are essential for the degradation of RNA in RNA-DNA hybrids. They are involved in various cellular processes, including DNA replication, repair, and recombination. These enzymes are found in both prokaryotes and eukaryotes and are crucial for maintaining genomic stability.

Nitrosomonas europaea and Its Genetic Engineering

Nitrosomonas europaea is a gram-negative bacterium known for its role in nitrification, converting ammonia into nitrite. It has been extensively studied for its metabolic pathways and genetic manipulation capabilities. For instance, N. europaea has been engineered to express the hemoglobin gene from Vitreoscilla, enhancing its oxygen uptake and ammonia oxidation capabilities .

Research Findings and Applications

While specific research findings on rnhA are lacking, studies on related enzymes and genetic modifications in N. europaea highlight the potential for enhancing metabolic processes through genetic engineering. For example, the expression of bacterial hemoglobin in N. europaea improved its respiratory abilities .

Data Table: Overview of Related Enzymes and Genetic Modifications

Enzyme/ModificationDescriptionSource
VHb in N. europaeaEnhances oxygen uptake and ammonia oxidation
Ribonuclease HII (rnhB)Produced in yeast for high purity
MazF EndoribonucleaseInvolved in stress response, targets specific RNA sequences

Future Directions

  • Genetic Engineering: Further studies on genetic modifications in N. europaea could reveal new applications for enzymes like rnhA.

  • Enzyme Characterization: Detailed biochemical characterization of rnhA would provide insights into its potential uses in biotechnology and molecular biology.

References

  1. Engineering of Nitrosomonas europaea to express Vitreoscilla hemoglobin. PMC4522006.

  2. Complete Genome Sequence of the Ammonia-Oxidizing Bacterium Nitrosomonas europaea. PMC154410.

  3. Transcriptomic Response of Nitrosomonas europaea Transitioned from Ammonia- to Oxygen-Limited Steady-State Growth. PMC6967387.

  4. Transcriptomic response of Nitrosomonas europaea transitioned from ammonia- to oxygen-limited steady-state growth. bioRxiv.

  5. Nitrosomonas europaea MazF Specifically Recognises the UGG Triplet. PMC6186784.

  6. Rhodococcus comparative genomics reveals a phylogenomic-dependent non-ribosomal peptide synthetase distribution. PMC8477407.

  7. Recombinant Nitrosomonas europaea Ribonuclease HII (rnhB). Cusabio.

  8. Transcription of All amoC Copies Is Associated with Recovery of Nitrosomonas europaea. Journal of Bacteriology.

  9. Genome analysis of secondary metabolite-biosynthetic gene clusters of Photorhabdus akhurstii subsp. akhurstii. PMC9491552.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact 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 consolidate the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference for your consideration.
Shelf Life
Shelf life depends on 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. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
rnhA; NE0140; Ribonuclease H; RNase H; EC 3.1.26.4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-161
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nitrosomonas europaea (strain ATCC 19718 / CIP 103999 / KCTC 2705 / NBRC 14298)
Target Names
rnhA
Target Protein Sequence
MQLEEGVKLV EIFTDGACKG NPGIGGWGVC LKFDGEVREF FGGEPVTTNN RMELLAAIRA LQALESLPDT GQSLRVQLHT DSQYVQKGIS EWVHSWKKRG WLTADKKPVK NEALWKELDQ LSRRYQVEWF WVRGHNGHDG NERADMLANR GVVSVLSEKA D
Uniprot No.

Target Background

Function
Endonuclease that specifically degrades RNA within RNA-DNA hybrid molecules.
Database Links

KEGG: neu:NE0140

STRING: 228410.NE0140

Protein Families
RNase H family
Subcellular Location
Cytoplasm.

Q&A

What is Nitrosomonas europaea Ribonuclease H (rnhA) and what is its primary function?

Nitrosomonas europaea Ribonuclease H (rnhA, gene designation NE0140) is an endonuclease that specifically degrades the RNA strand of RNA-DNA hybrids. The protein is encoded by a 486 bp gene located at position 165523-166008 on the positive strand of the N. europaea ATCC 19718 genome. RnhA belongs to the COG0328 functional category associated with DNA replication, recombination, and repair processes, and has the EC number 3.1.26.4 . Similar to other ribonucleases H, it likely functions in DNA replication by mediating the removal of RNA primers during lagging-strand synthesis (Okazaki fragment processing), a critical step in genomic DNA replication .

How does rnhA differ from other bacterial ribonucleases H?

Although the core catalytic function of ribonucleases H is conserved across species, N. europaea rnhA shows distinctive sequence characteristics. Sequence alignment analysis reveals that N. europaea rnhA shares approximately 52% identity with RnhA found in the tcd island of other bacteria . This moderate level of conservation suggests functional similarities while maintaining organism-specific adaptations. Structurally, the rnhA protein, like most type I RNases H, likely requires divalent metal ions (such as magnesium or manganese) as cofactors for catalytic activity, enabling the endonucleolytic cleavage of RNA to generate 5'-phosphomonoester products .

What expression systems are commonly used for producing recombinant N. europaea rnhA?

While the search results don't specifically detail expression systems for N. europaea rnhA, recombinant ribonucleases are typically expressed in E. coli systems. Based on analogous approaches with human RNASEH2A, effective expression involves cloning the rnhA gene into a vector containing a histidine tag to facilitate purification. The protein can then be purified through proprietary chromatographic techniques, producing a single, non-glycosylated polypeptide chain . For N. europaea proteins, researchers should consider codon optimization to account for potential codon usage bias between N. europaea and the expression host.

What are the recommended purification strategies for obtaining high-activity recombinant N. europaea rnhA?

For optimal purification of recombinant N. europaea rnhA, a multi-step chromatographic approach is recommended. Begin with immobilized metal affinity chromatography (IMAC) using Ni-NTA resins for initial capture of His-tagged rnhA. This should be followed by ion exchange chromatography to remove contaminants with different charge properties, and finally size exclusion chromatography to achieve high purity. Throughout purification, maintain buffers containing divalent metal ions (typically 5-10 mM MgCl₂ or MnCl₂) to stabilize the enzyme's active site. Activity can be preserved by adding reducing agents such as DTT (1-5 mM) and glycerol (10-20%) to storage buffers, with snap freezing in liquid nitrogen for long-term storage at -80°C.

The table below summarizes the recommended purification protocol:

Purification StepMethodBuffer CompositionKey Parameters
Initial CaptureIMAC (Ni-NTA)50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazoleGradual imidazole elution (10-250 mM)
Intermediate PurificationIon Exchange20 mM HEPES pH 7.5, 50-500 mM NaCl gradientpH optimized for rnhA (7.0-8.0)
PolishingSize Exclusion20 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl₂Flow rate 0.5 ml/min
Storage-20 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl₂, 1 mM DTT, 50% glycerolStore at -80°C in small aliquots

How can researchers effectively design mutation studies to investigate the catalytic mechanism of N. europaea rnhA?

To investigate the catalytic mechanism of N. europaea rnhA, researchers should focus on designing targeted mutations of conserved residues likely involved in catalysis or substrate binding. Based on sequence alignments with other RNases H and the knowledge of the protein's function in degrading RNA-DNA hybrids , the following approach is recommended:

  • Perform multiple sequence alignment with well-characterized RNases H to identify conserved residues, particularly those in the active site that coordinate metal ions or interact with the substrate.

  • Design site-directed mutagenesis experiments targeting:

    • Aspartic acid and glutamic acid residues that likely coordinate catalytic metal ions

    • Basic residues (lysine, arginine) that may interact with the phosphate backbone

    • Residues specific to N. europaea that might confer unique substrate preferences

  • Establish a reliable activity assay using fluorescently labeled RNA-DNA hybrid substrates to quantitatively measure the effects of each mutation.

  • Evaluate both kinetic parameters (kcat, Km) and metal ion preferences for each mutant to build a comprehensive model of the catalytic mechanism.

  • Complement biochemical studies with structural analyses (X-ray crystallography or cryo-EM) of wild-type and key mutant proteins to visualize changes in active site architecture.

What are the challenges in studying the in vivo role of rnhA in Nitrosomonas europaea?

Investigating the in vivo function of rnhA in N. europaea presents several significant challenges. First, N. europaea has a relatively slow growth rate compared to model organisms like E. coli, making genetic manipulation and phenotypic analysis time-consuming. Second, as an obligate chemolithoautotroph that oxidizes ammonia for energy, N. europaea requires specialized growth conditions with precise control of ammonia and oxygen concentrations .

For gene disruption studies, researchers must consider that rnhA may be essential for viability, as it plays a crucial role in DNA replication. If complete knockouts are lethal, conditional expression systems or partial knockdowns would be necessary. Additionally, the organism's complex response to environmental stressors, particularly oxygen limitation, can confound the interpretation of phenotypes resulting from rnhA manipulation .

Another challenge is the lack of standardized genetic tools for N. europaea compared to model organisms. Researchers would need to optimize transformation protocols, selection markers, and expression systems specifically for this organism. Complementation studies would be essential to confirm that observed phenotypes are directly attributed to rnhA disruption rather than polar effects or secondary mutations.

How can researchers integrate structural and functional studies of N. europaea rnhA?

A comprehensive research approach integrating structural and functional analyses of N. europaea rnhA should follow this methodological framework:

  • Structural determination: Solve the three-dimensional structure of rnhA using X-ray crystallography or cryo-EM, focusing on both apo-enzyme and substrate-bound forms. This provides the foundation for understanding the spatial arrangement of catalytic residues and substrate interactions.

  • Computational modeling: Perform molecular dynamics simulations to understand protein flexibility, substrate binding modes, and the role of metal ions in catalysis. This can predict key residues for subsequent functional validation.

  • Biochemical characterization: Systematically analyze substrate specificity using various RNA-DNA hybrid configurations to determine the optimal substrate characteristics (length, sequence preferences, structural features).

  • Structure-guided mutagenesis: Based on the structural data, design mutations targeting:

    • Catalytic residues

    • Substrate binding interface

    • Protein stability elements

    • Species-specific regions that differentiate N. europaea rnhA from other bacterial RNases H

  • In vivo functional correlation: Develop genetic complementation systems where mutant variants of rnhA are expressed in rnhA-deficient backgrounds to correlate biochemical properties with physiological function.

  • Interaction partner identification: Employ pull-down assays coupled with mass spectrometry to identify proteins that interact with rnhA in N. europaea, potentially revealing its integration into larger DNA metabolism complexes.

This integrated approach allows researchers to connect atomic-level structural insights with cellular functions, providing a comprehensive understanding of how rnhA contributes to N. europaea biology.

What is the relationship between rnhA activity and nitrogen metabolism in Nitrosomonas europaea?

The connection between rnhA activity and nitrogen metabolism in N. europaea represents an intriguing but unexplored research area. While no direct evidence links rnhA to nitrogen metabolism, several hypotheses warrant investigation:

First, as an ammonia-oxidizing bacterium, N. europaea undergoes substantial transcriptional changes under different growth conditions, particularly in response to oxygen limitation . During oxygen-limited growth, genes involved in nitrification and denitrification show altered expression patterns . Given rnhA's role in DNA replication and repair, changes in growth conditions that affect cell division rates would indirectly impact the requirement for rnhA activity.

Second, the stress response associated with reactive nitrogen species produced during ammonia oxidation may increase DNA damage, potentially elevating the importance of DNA repair pathways that might involve rnhA. Under oxygen limitation, N. europaea increases production of nitric oxide (NO) and nitrous oxide (N₂O) , which could lead to nitrosative stress affecting DNA integrity.

Third, the coordination of nitrogen metabolism with DNA replication in slow-growing bacteria like N. europaea may involve regulatory mechanisms that orchestrate these processes, potentially involving post-transcriptional regulation where rnhA activity could be relevant.

To investigate these potential relationships, researchers could examine:

  • rnhA expression levels under different nitrogen and oxygen regimes

  • Effects of rnhA overexpression or depletion on nitrogen transformation rates

  • Potential protein-protein interactions between rnhA and nitrogen metabolism enzymes

  • Correlation between DNA replication rates and ammonia oxidation activity

How should researchers design activity assays for recombinant N. europaea rnhA?

For robust activity assays of recombinant N. europaea rnhA, researchers should implement the following methodological approach:

  • Substrate preparation: Synthesize model RNA-DNA hybrid substrates with varying features:

    • Length: 10-30 bp hybrids to determine size preferences

    • Sequence composition: Vary GC content to test sequence bias

    • Structure: Include both linear and structured (hairpin-containing) hybrids

    • Modifications: Fluorescently labeled substrates for real-time monitoring

  • Assay conditions optimization:

    • Buffer composition: Test various pH ranges (6.5-9.0) and ionic strengths

    • Metal cofactors: Compare activity with Mg²⁺, Mn²⁺, and other divalent cations at 1-10 mM

    • Temperature: Determine optimal temperature and stability (25-55°C range)

    • Enzyme concentration: Establish linear range for activity measurements

  • Detection methods:

    • Gel-based assays: Denaturing PAGE with radiolabeled or fluorescent substrates

    • Real-time fluorescence assays: FRET-based systems for continuous monitoring

    • High-throughput formats: Microplate-based assays with quenched fluorescent substrates

  • Kinetic analysis:

    • Determine Km, kcat, and kcat/Km under optimized conditions

    • Evaluate product release patterns to establish processivity vs. distributive action

    • Assess inhibition profiles with various ions and nucleic acid structures

  • Controls and validation:

    • Include commercially available RNases H as positive controls

    • Use catalytically inactive mutants as negative controls

    • Verify that activity is specific to RNA-DNA hybrids vs. dsRNA or dsDNA

What approaches can be used to study the physiological role of rnhA in N. europaea under different environmental conditions?

To investigate rnhA's physiological role in N. europaea under varying environmental conditions, researchers should employ a multi-faceted approach:

  • Transcriptomic profiling:

    • Conduct RNA-seq analyses comparing rnhA expression across growth conditions (ammonia-limited vs. oxygen-limited cultures)

    • Correlate rnhA expression patterns with genes involved in DNA replication and nitrogen metabolism

    • Analyze promoter elements to identify potential regulatory factors

  • Genetic manipulation strategies:

    • Develop conditional knockdown systems (inducible antisense RNA or CRISPRi)

    • Create strains with tagged rnhA (e.g., FLAG-tag) for localization and interaction studies

    • Design reporter fusions to monitor rnhA expression in real-time

  • Physiological characterization:

    • Measure growth rates, ammonia oxidation, and nitrite production in rnhA-modulated strains

    • Analyze DNA replication rates using incorporation of labeled nucleotides

    • Assess RNA primer accumulation on genomic DNA

  • Stress response studies:

    • Test sensitivity to DNA-damaging agents in rnhA-modified strains

    • Evaluate responses to oxidative and nitrosative stress

    • Analyze survival under nutrient limitation and pH stress

  • Microscopy approaches:

    • Utilize fluorescence microscopy with labeled rnhA to track subcellular localization

    • Implement time-lapse imaging to correlate rnhA positioning with cell cycle stages

    • Apply super-resolution techniques to visualize colocalization with replication machinery

By combining these approaches, researchers can establish connections between rnhA function and N. europaea's adaptation to different environmental conditions, particularly the transitions between ammonia-limited and oxygen-limited states that significantly impact its nitrogen metabolism .

How should researchers interpret conflicting results from in vitro versus in vivo studies of rnhA function?

When faced with discrepancies between in vitro biochemical data and in vivo functional studies of N. europaea rnhA, researchers should follow this systematic approach to reconciliation:

  • Evaluate experimental conditions:

    • Assess whether in vitro conditions (pH, salt concentration, metal cofactors) appropriately mimic the intracellular environment of N. europaea

    • Consider that N. europaea faces unique physiological challenges as an ammonia-oxidizing bacterium, including exposure to reactive nitrogen species

  • Consider protein interactions:

    • In vivo, rnhA likely functions within protein complexes that may modify its activity

    • Identify potential interaction partners through pull-down assays or bacterial two-hybrid systems

    • Test whether adding cellular extracts modifies in vitro activity

  • Examine substrate accessibility:

    • The concentration and nature of RNA-DNA hybrids may differ significantly between test tube and cellular environments

    • In vivo, competing nucleic acid binding proteins may restrict rnhA access to substrates

  • Account for compensatory mechanisms:

    • Redundant enzymatic activities may mask phenotypes in vivo

    • Search the N. europaea genome for alternative RNase H activities or other enzymes that process RNA-DNA hybrids

  • Quantitative considerations:

    • Calculate whether the measured in vitro activity rates are sufficient to support cellular requirements

    • Develop mathematical models integrating enzyme kinetics with cellular replication rates

  • Develop bridging experiments:

    • Design reconstituted systems of increasing complexity to transition between pure in vitro and complete in vivo conditions

    • Use permeabilized cells to allow controlled substrate access while maintaining cellular architecture

This methodical approach helps researchers identify the specific factors contributing to discrepancies and develop a more comprehensive understanding of rnhA function in its native context.

What computational approaches can help predict substrate specificity of N. europaea rnhA?

Computational methods offer powerful tools for predicting the substrate specificity of N. europaea rnhA. A comprehensive computational strategy should include:

  • Homology modeling and structural prediction:

    • Generate 3D structural models based on crystal structures of related RNases H

    • Refine models using molecular dynamics simulations in explicit solvent

    • Validate model quality using tools like PROCHECK and MolProbity

  • Binding site analysis:

    • Identify conserved catalytic residues through multiple sequence alignments

    • Map electrostatic surface potentials to predict nucleic acid binding regions

    • Apply computational solvent mapping to identify hot spots for molecular recognition

  • Substrate docking simulations:

    • Generate models of various RNA-DNA hybrid configurations

    • Perform molecular docking using tools like AutoDock or HADDOCK

    • Calculate binding energies and identify key interaction residues

  • Molecular dynamics-based approaches:

    • Conduct MD simulations of enzyme-substrate complexes to assess binding stability

    • Implement enhanced sampling techniques (metadynamics, umbrella sampling) to estimate free energy landscapes

    • Model metal ion coordination and its impact on substrate positioning

  • Machine learning integration:

    • Train models using existing RNase H specificity data across species

    • Implement feature extraction from sequence and structural data

    • Develop classifiers to predict optimal substrate characteristics

  • Validation procedures:

    • Design in silico mutagenesis to test computational predictions

    • Compare predictions with experimental data from related RNases H

    • Iteratively refine models based on experimental feedback

These computational approaches generate testable hypotheses about N. europaea rnhA substrate preferences, guiding the design of tailored substrates for experimental validation and potentially revealing unique specificities related to N. europaea's ecological niche.

What are common problems in expressing functional recombinant N. europaea rnhA and how can they be addressed?

Researchers frequently encounter challenges when expressing recombinant N. europaea rnhA. The following table summarizes common problems and their solutions:

ProblemPotential CausesSolutions
Low expression yieldCodon bias between N. europaea and expression hostOptimize codons for expression host; use specialized strains with rare tRNAs
Toxicity to host cellsUse tightly regulated inducible promoters; lower growth temperature
mRNA secondary structureModify 5' region of gene to reduce secondary structure; use fusion tags
Insoluble protein/inclusion bodiesImproper foldingCo-express with chaperones; lower induction temperature to 16-18°C
Hydrophobic patches exposureAdd solubilizing fusion partners (SUMO, MBP, TrxA)
Improper disulfide formationExpress in specialized strains (SHuffle, Origami)
Low activity of purified proteinMetal cofactor issuesInclude appropriate divalent cations (Mg²⁺, Mn²⁺) in all buffers
Oxidation of critical residuesAdd reducing agents (DTT, β-mercaptoethanol)
Improper pH for folding/activityTest buffer conditions systematically
Protein instabilityProteolytic degradationInclude protease inhibitors; use protease-deficient strains
Aggregation tendenciesAdd stabilizing agents like glycerol or arginine
Thermal instabilityMaintain at 4°C throughout purification; avoid freeze-thaw cycles

Additionally, researchers should consider the cellular environment of N. europaea when designing expression and purification strategies. As an ammonia-oxidizing bacterium with a different intracellular milieu than typical expression hosts, N. europaea proteins may require specific conditions to maintain their native structure and activity .

How can researchers distinguish between the activity of rnhA and other ribonucleases in complex samples?

Distinguishing rnhA activity from other ribonucleases in complex biological samples requires strategic experimental design:

  • Substrate specificity exploitation:

    • Design assays using RNA-DNA hybrid substrates specifically cleaved by RNase H

    • Include control substrates (ssRNA, dsRNA) that should not be cleaved by rnhA but might be targeted by other ribonucleases

    • Use structural variants of RNA-DNA hybrids to identify signature cleavage patterns

  • Inhibitor profiling:

    • Employ specific RNase H inhibitors (e.g., hydroxylaminopurines)

    • Test sensitivity to general RNase inhibitors (RNasin, RNase OUT)

    • Create an inhibition profile using various metal chelators and ion concentrations

  • Immunodepletion approaches:

    • Develop specific antibodies against N. europaea rnhA

    • Perform immunodepletion of complex samples to remove rnhA

    • Compare activity profiles before and after depletion

  • Genetic approaches:

    • Use samples from rnhA-depleted or overexpressing strains

    • Complement rnhA-deficient samples with recombinant protein

    • Employ CRISPR/Cas9 knockdown in appropriate systems

  • Biochemical fractionation:

    • Separate activities using ion exchange or size exclusion chromatography

    • Track rnhA using activity assays and western blotting across fractions

    • Identify co-purifying ribonucleases by mass spectrometry

  • Kinetic discrimination:

    • Determine reaction kinetics under varying pH and salt conditions

    • Exploit differences in temperature sensitivity

    • Measure activity in the presence of specific metal ions

What emerging technologies could advance our understanding of N. europaea rnhA function and regulation?

Several cutting-edge technologies show promise for deepening our understanding of N. europaea rnhA:

  • CRISPR-based approaches:

    • CRISPRi for tunable gene repression to create partial knockdowns

    • CRISPR-mediated base editing for precise mutagenesis without double-strand breaks

    • CRISPR screening to identify genetic interactions with rnhA

  • Advanced imaging technologies:

    • Super-resolution microscopy to visualize rnhA localization during cell cycle progression

    • Live-cell single-molecule tracking to monitor rnhA dynamics

    • Correlative light and electron microscopy to connect rnhA positioning with cellular ultrastructure

  • Structural biology innovations:

    • Cryo-electron tomography of whole cells to visualize rnhA in native context

    • Time-resolved X-ray crystallography to capture catalytic intermediates

    • Hydrogen-deuterium exchange mass spectrometry to map protein dynamics

  • Next-generation sequencing applications:

    • RNA-DNA hybrid sequencing (DRIP-seq) to map hybrid accumulation in rnhA mutants

    • Nascent strand sequencing to analyze replication defects

    • High-throughput mutagenesis coupled with deep sequencing for structure-function studies

  • Systems biology integration:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics

    • Flux balance analysis to model the impact of rnhA on cellular metabolism

    • Network analysis to position rnhA within the broader cellular response to environmental stressors

  • Synthetic biology tools:

    • Engineered allosteric riboswitches to control rnhA expression

    • Orthogonal translation systems for site-specific incorporation of non-canonical amino acids

    • Cell-free expression systems to study rnhA in controlled biochemical environments

These technologies will enable researchers to move beyond traditional biochemical characterization toward understanding rnhA's dynamic role in the complex physiology of N. europaea, particularly under changing environmental conditions relevant to its ecological niche .

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.