Recombinant Nitrosomonas europaea Exodeoxyribonuclease 7 small subunit (xseB)

Shipped with Ice Packs
In Stock

Description

Molecular Characterization of Recombinant xseB

Recombinant xseB is produced via heterologous expression in Escherichia coli, ensuring high purity (>85% by SDS-PAGE) and stability. Key molecular details include:

PropertySpecification
Gene NamexseB
Uniprot IDQ82VD5
Source OrganismNitrosomonas europaea (strain ATCC 19718)
Recombinant Expression HostE. coli
Protein Length112 amino acids
Sequence (Partial)MRKKSSSNKEETALHPPPENFETATAELEQIVAGMETGQMSLEDALSAY... (full sequence in )
Storage Stability6 months (liquid, -20°C/-80°C); 12 months (lyophilized, -20°C/-80°C)

This subunit forms part of the Exodeoxyribonuclease VII (ExoVII) complex, which degrades single-stranded DNA (ssDNA) during DNA repair .

Biological Role in Nitrosomonas europaea

xseB contributes to genome maintenance in N. europaea, a chemolithoautotroph reliant on ammonia oxidation. Key findings:

  • DNA Repair Mechanism: ExoVII (comprising xseB and xseA) resolves ssDNA gaps and removes mutagenic lesions, critical for surviving oxidative stress from ammonia metabolism .

  • Genomic Adaptations: The N. europaea genome encodes 2,460 proteins, with xseB part of a minimal DNA repair toolkit tailored for its energy-intensive lithotrophic lifestyle .

  • Stress Response: Transcriptomic studies show upregulation of DNA repair genes under oxygen limitation, suggesting xseB’s role in mitigating DNA damage during nitrosative stress .

Applications in Biotechnology

Recombinant xseB is utilized in:

  • Molecular Biology: Tool for ssDNA degradation in cloning workflows .

  • Bioremediation Studies: N. europaea’s DNA repair pathways are engineered to withstand pollutants (e.g., chlorinated hydrocarbons), with xseB as a biomarker for stress response .

  • Enzyme Engineering: Optimized recombinant production enables structural studies for industrial enzyme design .

Research Gaps and Future Directions

  • Mechanistic Studies: The precise role of xseB in N. europaea’s niche environments (e.g., wastewater biofilms) remains underexplored.

  • Biochemical Assays: Kinetic parameters (e.g., KmK_m, VmaxV_{max}) for the recombinant enzyme are yet to be published.

  • Synergy with AMO Enzymes: Potential crosstalk between xseB and ammonia monooxygenase (AMO) during pollutant degradation warrants investigation .

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 settle 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%, which can serve as a reference for your preparation.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and 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
Tag type is 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
xseB; NE1159Exodeoxyribonuclease 7 small subunit; EC 3.1.11.6; Exodeoxyribonuclease VII small subunit; Exonuclease VII small subunit
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-89
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nitrosomonas europaea (strain ATCC 19718 / CIP 103999 / KCTC 2705 / NBRC 14298)
Target Names
xseB
Target Protein Sequence
MRKKSSSNKE ETALHPPPEN FETATAELEQ IVAGMETGQM SLEDALSAYK RGVELLQYCQ NILKNSQQQI KILEADMLKH FSPAEHDAS
Uniprot No.

Target Background

Function
This protein bidirectionally degrades single-stranded DNA into larger, acid-insoluble oligonucleotides, which are subsequently further degraded into smaller, acid-soluble oligonucleotides.
Database Links

KEGG: neu:NE1159

STRING: 228410.NE1159

Protein Families
XseB family
Subcellular Location
Cytoplasm.

Q&A

What is the basic structure of Exodeoxyribonuclease VII and how does the xseB subunit contribute to its function?

Exodeoxyribonuclease VII (ExoVII) is a bacterial nuclease complex involved in DNA repair and recombination that specifically hydrolyzes single-stranded DNA. The complex comprises two types of subunits: a large subunit XseA (51.8 kDa) and a small subunit XseB (10.5 kDa), encoded by the xseA and xseB genes, respectively . The quaternary structure of the functional complex is estimated to contain one XseA subunit and four XseB subunits based on densitometric analysis of protein bands in Coomassie-stained polyacrylamide gels .

The XseA subunit contains four distinct structural domains: an N-terminal OB-fold domain responsible for DNA binding, a middle putative catalytic domain containing the active site residues (D155, R205, H238, and D241), a coiled-coil domain, and a short C-terminal segment . The XseB subunit appears to interact with the coiled-coil domain of XseA, with multiple XseB molecules binding to a single XseA molecule . While the catalytic activity resides in the XseA subunit, the XseB subunits are crucial for stabilizing the complex and potentially modulating its activity or substrate specificity.

What are the optimal conditions for recombinant expression of N. europaea xseB in E. coli expression systems?

The recombinant expression of N. europaea xseB in E. coli expression systems can be optimized using protocols adapted from successful expressions of similar proteins. Based on methodologies used for E. coli ExoVII components, the following protocol can be recommended:

For vector construction, clone the N. europaea xseB gene into a pET-based expression vector with an N-terminal His6-tag for purification purposes, similar to the approach used for E. coli xseB in the ASKA library . The promoter region should be carefully selected, with the T7 promoter system being advantageous for controlled, high-level expression.

Expression conditions:

  • Transform the construct into E. coli BL21(DE3) or similar expression strains.

  • Grow transformed cells in LB medium supplemented with appropriate antibiotics at 37°C until OD600 reaches 0.6-0.8.

  • Induce protein expression with 0.5-1 mM IPTG.

  • Lower the temperature to 16-25°C post-induction to enhance proper folding.

  • Continue expression for 16-20 hours.

These conditions help prevent the formation of inclusion bodies while maximizing soluble protein yield. Additionally, co-expression with the XseA subunit may improve XseB stability and solubility, as these proteins naturally function as a complex. For highest activity, consider using an E. coli strain with knockout mutations in the native xseA and xseB genes (similar to the Δ xseA and Δ xseB strains used in previous studies) to prevent contamination with host proteins .

What purification strategies yield the highest purity and activity of recombinant N. europaea XseB?

A multi-step purification strategy is recommended to obtain high-purity, active recombinant N. europaea XseB. Based on approaches used for similar nucleases, the following protocol can be implemented:

  • Initial capture using affinity chromatography: Utilize the His6-tag by loading the clarified cell lysate onto a Ni-NTA column. Wash with buffer containing 20-50 mM imidazole to remove weakly bound contaminants, then elute the target protein with 250-300 mM imidazole. This step typically yields protein of approximately 70-80% purity.

  • Ion exchange chromatography: Apply the partially purified protein to an anion exchange column (e.g., Q Sepharose) after dialysis to remove imidazole. Perform elution using a linear NaCl gradient (0-500 mM). This step separates the target protein from contaminants with different charge properties.

  • Size exclusion chromatography: As a final polishing step, apply the sample to a gel filtration column (e.g., Superdex 75) to separate monomeric XseB from aggregates and remaining contaminants. This step also allows buffer exchange to the storage buffer.

For assessing purity at each stage, SDS-PAGE with Coomassie staining is recommended, with Western blotting using anti-His antibodies for confirmation of identity. Activity can be measured using a single-stranded DNA degradation assay, similar to those employed for E. coli ExoVII .

The table below summarizes the expected outcomes at each purification stage:

Purification StageExpected PurityTypical Yield (% of starting material)Quality Control Methods
Ni-NTA Affinity70-80%60-70%SDS-PAGE, Western blot
Ion Exchange85-90%40-50%SDS-PAGE, activity assay
Size Exclusion>95%30-40%SDS-PAGE, activity assay, mass spectrometry

For optimal storage, the purified protein should be kept in buffer containing 50 mM Tris-HCl (pH 8.0), 150 mM NaCl, 1 mM DTT, and 10% glycerol at -80°C. Aliquoting before freezing prevents repeated freeze-thaw cycles that can decrease activity.

How can researchers effectively design experiments to study the interaction between XseA and XseB subunits in N. europaea?

To effectively study the interactions between XseA and XseB subunits in N. europaea, researchers should implement a multi-faceted approach combining biochemical, biophysical, and genetic techniques:

  • Co-immunoprecipitation (Co-IP): Express tagged versions of XseA and XseB (e.g., His-tagged XseA and FLAG-tagged XseB) in a heterologous system or in N. europaea. Perform pull-down assays to confirm physical interaction and identify the stoichiometry of the complex. This approach can verify the reported 1:4 ratio of XseA:XseB observed in E. coli .

  • Yeast two-hybrid or bacterial two-hybrid assays: These genetic systems can map the specific domains involved in the XseA-XseB interaction. Based on E. coli studies, the coiled-coil domain of XseA is likely involved in binding multiple copies of XseB .

  • Site-directed mutagenesis: Generate targeted mutations in potential interaction interfaces, particularly in the coiled-coil domain of XseA and complementary regions of XseB. Evaluate the effects of these mutations on complex formation and enzyme activity.

  • Analytical ultracentrifugation and size-exclusion chromatography coupled with multi-angle light scattering (SEC-MALS): These techniques provide information about the molecular weight and stoichiometry of the complex under native conditions.

  • Microscale thermophoresis (MST) or isothermal titration calorimetry (ITC): These methods can determine binding affinities and thermodynamic parameters of the XseA-XseB interaction.

  • Structural studies: If resources permit, X-ray crystallography or cryo-electron microscopy of the complex would provide detailed structural insights into the interaction interfaces.

Controls should include:

  • Individual subunits expressed and analyzed separately

  • Known non-interacting proteins to establish specificity

  • Comparative analyses with E. coli ExoVII components to identify species-specific interactions

For data analysis, computational methods such as molecular dynamics simulations can complement experimental findings by predicting interaction dynamics and potential conformational changes upon binding.

What approaches can differentiate the catalytic contribution of XseB in the ExoVII complex from its structural role?

Differentiating between the catalytic and structural roles of the XseB subunit in the ExoVII complex requires sophisticated experimental designs that systematically isolate these functions. Based on what we know about E. coli ExoVII, the following approaches are recommended:

  • Structure-function analysis through targeted mutations: Create a series of XseB mutants with alterations in conserved residues. Test these mutants in reconstituted ExoVII complexes for:

    • Ability to form complexes with XseA (structural role)

    • Nuclease activity of the resulting complexes (catalytic contribution)

    Mutations that disrupt complex formation but preserve the structure of XseB (as confirmed by circular dichroism) would indicate regions involved in the structural role. Mutations that allow complex formation but reduce activity would suggest catalytic contributions.

  • Complementation studies in knockout strains: Using Δ xseB strains similar to those from the Keio collection , perform complementation with:

    • Wild-type XseB

    • Truncated XseB variants

    • XseB with point mutations

    Measure both complex formation and nuclease activity to correlate specific XseB regions with either function.

  • Minimal functional domain analysis: Create a series of XseB truncations to identify the minimal region required for:

    • Binding to XseA

    • Supporting catalytic activity of the complex

    These experiments can determine if separate domains exist for structural versus catalytic functions.

  • Cross-linking studies coupled with mass spectrometry: This approach can identify specific residues of XseB that interact with XseA or with DNA substrates, providing spatial information about the potential catalytic contributions of XseB.

  • Single-molecule studies: Using techniques like FRET (Fluorescence Resonance Energy Transfer), examine conformational changes in the complex during catalysis, potentially revealing dynamic roles of XseB in the reaction mechanism.

Data from these experiments should be integrated with bioinformatic analyses, including sequence conservation patterns across bacterial species and structural predictions, to build a comprehensive model of XseB's dual roles in the ExoVII complex.

How can recombinant N. europaea expressing XseB be utilized as a biosensor for environmental contaminants?

Recombinant N. europaea can be engineered as an effective biosensor for environmental contaminants by leveraging the stress response pathways that regulate xseB expression. This approach builds upon successful precedents with other genes in N. europaea, such as the mbla and clpB genes, which have been used to create biosensors for chloroform and other stressors .

Methodology for developing an xseB-based biosensor:

  • Promoter identification and characterization: Identify the promoter region of the xseB gene in N. europaea and characterize its response to various environmental stressors, particularly those that cause DNA damage, which likely activates the ExoVII DNA repair pathway.

  • Reporter fusion construction: Create a transcriptional fusion between the xseB promoter and a reporter gene such as gfp (green fluorescent protein) similar to the pPRO/mbla4 and pPRO/clpb7 constructs described for chloroform sensing . The resulting construct would express GFP when the xseB promoter is activated by environmental stressors.

  • Transformation into N. europaea: Transform N. europaea with the xseB promoter-gfp fusion construct using established transformation protocols .

  • Calibration with known DNA-damaging agents: Expose the transformed N. europaea to various concentrations of known DNA-damaging agents (e.g., UV radiation, hydrogen peroxide, DNA-targeting antibiotics) and measure the resulting GFP fluorescence. This establishes a dose-response curve and determines the sensitivity and detection range of the biosensor.

  • Validation with environmental samples: Test the biosensor with environmental samples containing suspected DNA-damaging contaminants, comparing the response to the established calibration curves.

The advantage of using xseB-based biosensors is their potential specificity for contaminants that cause DNA damage, particularly to single-stranded DNA, which is the substrate for ExoVII. This approach could complement existing biosensors based on other stress response genes, providing a more comprehensive contaminant detection system.

Based on data from similar biosensor systems in N. europaea, the following performance metrics could be expected:

ContaminantDetection RangeResponse TimeFold Increase in Fluorescence
Chloroform7-100 μM4-6 hours3-18 fold
H₂O₂2.5-7.5 mM2-4 hours8-10 fold
UV radiation5-50 J/m²1-2 hours5-15 fold
Heavy metals0.1-2 mM6-8 hours2-7 fold

These values are extrapolated from the performance of mbla and clpB promoter-based biosensors and would need to be experimentally verified for an xseB-based system.

What are the experimental considerations when studying the expression of xseB in N. europaea under different environmental stressors?

When studying the expression of xseB in N. europaea under various environmental stressors, several critical experimental considerations must be addressed to ensure reliable and reproducible results:

  • Growth conditions and physiological state standardization: N. europaea has specific growth requirements as an ammonia-oxidizing bacterium. Maintain consistent ammonia concentrations, pH (typically 7.5-8.0), temperature (28-30°C), and dissolved oxygen levels across experiments. The physiological state of the cells significantly impacts stress responses, so standardize growth phase (typically mid-logarithmic) for all experiments .

  • Stressor application protocols: For each environmental stressor:

    • Chemical stressors: Prepare stock solutions fresh before each experiment; verify concentrations analytically when possible

    • Physical stressors (temperature, pH): Use gradual transitions to avoid shock responses

    • Radiation: Calibrate UV sources regularly; ensure uniform exposure across samples

  • Expression measurement methodologies:

    • RT-qPCR: Design primers specific to N. europaea xseB with validation for specificity and efficiency

    • Reporter systems: If using promoter-reporter fusions (e.g., xseB promoter-gfp), normalize fluorescence to cell density

    • Protein levels: Develop specific antibodies for XseB or use tagged versions for Western blotting

    • Transcriptomics: For global expression analysis, ensure adequate sequencing depth and appropriate normalization

  • Controls and normalization: Include both:

    • Untreated controls maintained under identical conditions

    • Housekeeping genes (e.g., 16S rRNA, rpoB) for RT-qPCR normalization

    • Standard curves with purified GFP for fluorescence quantification

  • Time-course considerations: As demonstrated with chloroform and hydrogen peroxide exposure experiments in N. europaea , stress responses can be highly time-dependent. Design experiments to capture both:

    • Acute responses (minutes to hours)

    • Adaptive responses (hours to days)

  • Multi-omics integration: To understand the biological context of xseB regulation:

    • Correlate transcriptomic data with proteomic measurements

    • Monitor enzymatic activity of ExoVII under stressors

    • Assess physiological parameters (e.g., growth rate, ammonia oxidation rate)

  • Statistical design and analysis:

    • Use at least 3-5 biological replicates for each condition

    • Apply appropriate statistical tests (ANOVA with post-hoc tests for multiple comparisons)

    • Consider using DOE (Design of Experiments) approach for multi-factor experiments

A key challenge is distinguishing direct effects on xseB expression from indirect effects mediated through general stress responses or growth inhibition. This requires careful experimental design with appropriate controls and potentially the use of mutant strains with altered stress response pathways.

How can CRISPR-Cas9 genome editing be optimized for modifying the xseB gene in N. europaea?

Optimizing CRISPR-Cas9 genome editing for modifying the xseB gene in N. europaea requires addressing several challenges specific to this ammonia-oxidizing bacterium, including its relatively slow growth rate, limited genetic tools, and unique physiological characteristics. The following comprehensive protocol addresses these challenges:

  • sgRNA design and validation:

    • Design multiple sgRNAs targeting different regions of the xseB gene using algorithms that account for the high GC content of N. europaea genome

    • Test sgRNA efficiency in vitro using purified Cas9 protein and PCR-amplified xseB target regions

    • Prioritize sgRNAs targeting non-conserved regions to minimize off-target effects

  • Delivery system optimization:

    • Construct a broad-host-range vector containing:

      • A codon-optimized Cas9 gene under a promoter active in N. europaea

      • The sgRNA expression cassette

      • Homology arms (800-1000 bp) flanking the desired modification site

      • A selectable marker compatible with N. europaea (e.g., kanamycin resistance)

    • Consider using a temperature-sensitive origin of replication for plasmid curing after successful editing

  • Transformation protocol:

    • Prepare electrocompetent N. europaea cells harvested at mid-logarithmic phase

    • Use electroporation with optimized parameters: 2.5 kV, 200 Ω, 25 μF

    • Allow extended recovery (24-48 hours) in ammonia-containing medium without selective pressure

    • Gradually introduce the selective agent to minimize stress

  • Screening and verification:

    • Design PCR primers flanking the modification site for rapid screening

    • Confirm modifications by sequencing

    • Verify the absence of off-target modifications by whole-genome sequencing of selected clones

    • Assess expression levels and functionality of the modified xseB gene

  • Phenotypic characterization:

    • Compare growth rates between wild-type and modified strains

    • Assess DNA repair capacity using DNA-damaging agents

    • Evaluate ammonia oxidation rates to ensure central metabolism is not affected

This approach can be used to generate various xseB modifications, including:

  • Complete gene deletions to study essentiality

  • Point mutations to investigate specific residues involved in XseA interaction

  • Promoter modifications to alter expression levels

  • Introduction of tags for protein localization studies

For researchers working with N. europaea, it is important to note that the editing efficiency is likely to be lower than in model organisms like E. coli, necessitating screening of a larger number of transformants and possibly multiple rounds of optimization.

What experimental approaches can reveal the role of XseB in N. europaea's response to oxidative stress from ammonia oxidation?

Investigating the role of XseB in N. europaea's response to oxidative stress from ammonia oxidation requires a multi-faceted approach that combines genetic manipulation, physiological assessments, and molecular analyses:

  • Generation of xseB mutant strains:

    • Create an xseB knockout strain using CRISPR-Cas9 or traditional homologous recombination

    • Develop complementation strains expressing wild-type xseB under native and inducible promoters

    • Engineer strains expressing tagged XseB for localization and interaction studies

  • Oxidative stress induction protocols:

    • Physiological stress: Vary ammonia concentrations (1-30 mM) to modulate endogenous ROS production

    • Exogenous stress: Apply H₂O₂ (2.5-7.5 mM) or other oxidants

    • Combined stressors: Test ammonia oxidation under chlorinated compound exposure (7-100 μM chloroform)

  • Comparative physiological assessments:

    • Measure growth rates and cell viability under oxidative stress conditions

    • Quantify ammonia oxidation rates using ion chromatography

    • Assess membrane integrity and energetics using fluorescent probes

    • Measure intracellular ROS levels using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA)

  • Molecular and biochemical analyses:

    • Quantify oxidative DNA damage using techniques like comet assay and 8-oxoG measurement

    • Monitor xseB transcription under various oxidative stress conditions using RT-qPCR

    • Assess XseB protein levels and modifications using Western blotting

    • Measure ExoVII enzymatic activity using single-stranded DNA degradation assays

    • Characterize protein-protein interactions using co-immunoprecipitation and mass spectrometry

  • Transcriptome and proteome analysis:

    • Perform RNA-seq to identify genes differentially expressed in xseB mutants under oxidative stress

    • Compare proteome profiles between wild-type and xseB mutant strains

    • Analyze post-translational modifications, particularly oxidative modifications

  • In vivo DNA repair assessment:

    • Develop reporter systems to quantify DNA repair efficiency

    • Measure mutation rates under various oxidative stress conditions

    • Assess recombination frequency as an indicator of DNA repair pathway activation

Based on transcriptomic data from N. europaea under oxygen limitation and other stressors , we can anticipate potential interactions between XseB and other components of stress response systems. The table below summarizes these potential interactions and corresponding experimental approaches:

Potential Interaction/PathwayExperimental ApproachExpected Outcome in xseB Mutants
Cytochrome c552 (cycB) pathwayCytochrome activity assaysAltered electron transfer efficiency
Nitrosocyanin (ncyA) expressionRT-qPCR, Western blotModified expression under O₂ limitation
NirK-dependent nitrite reductionNO and N₂O measurementChanged nitrogen oxide production
Cytochrome P460 (cytL) pathwayN₂O production assaysAltered hydroxylamine detoxification
Cytochrome c=-beta (cytS) expressionTranscriptome analysisModified N-oxide metabolism

This comprehensive approach will provide insights into the specific role of XseB in protecting N. europaea from oxidative DNA damage during ammonia oxidation, potentially revealing novel connections between DNA repair mechanisms and energy metabolism in this environmentally important bacterium.

What are the common challenges in achieving functional expression of recombinant N. europaea XseB and how can they be addressed?

Researchers frequently encounter several challenges when attempting to express functional recombinant N. europaea XseB. These challenges and their solutions are detailed below:

  • Poor solubility and inclusion body formation:

    • Problem: XseB may form inclusion bodies when overexpressed in E. coli, particularly at high induction temperatures.

    • Solutions:

      • Lower induction temperature to 16-18°C and extend expression time (16-24 hours)

      • Reduce IPTG concentration to 0.1-0.3 mM for gentler induction

      • Use solubility-enhancing fusion tags (SUMO, MBP, or TrxA) instead of simple His-tags

      • Co-express with molecular chaperones (GroEL/ES, DnaK/J) to assist proper folding

      • Consider co-expression with XseA, as the natural binding partner may enhance XseB stability

  • Proteolytic degradation:

    • Problem: Recombinant XseB may be subject to proteolytic degradation during expression or purification.

    • Solutions:

      • Use protease-deficient E. coli strains (e.g., BL21(DE3) pLysS)

      • Add protease inhibitors (PMSF, EDTA, Protease Inhibitor Cocktail) during all purification steps

      • Optimize buffer conditions (pH, salt concentration) to minimize protease activity

      • Consider C-terminal His-tags if N-terminal degradation occurs

  • Lack of catalytic activity:

    • Problem: Purified XseB may lack activity due to improper folding or absence of its XseA partner.

    • Solutions:

      • Ensure proper disulfide bond formation if applicable (check for reducing agents in buffers)

      • Co-purify with XseA to form the functional ExoVII complex

      • Verify protein structure using circular dichroism or thermal shift assays

      • Test activity under various buffer conditions, including different metal ions

  • Low expression yields:

    • Problem: Expression levels of N. europaea proteins in E. coli may be low due to codon usage differences.

    • Solutions:

      • Optimize codon usage for E. coli without altering the amino acid sequence

      • Use E. coli strains supplemented with rare tRNAs (e.g., Rosetta, CodonPlus)

      • Test different expression vectors with stronger promoters

      • Optimize media composition (consider auto-induction media)

  • Toxicity to host cells:

    • Problem: Expression of functional nucleases can be toxic to host cells.

    • Solutions:

      • Use tight expression control systems (T7-lac, araBAD) to minimize leaky expression

      • Express in E. coli strains that are knockout for endogenous xseA and xseB

      • Express an inactive mutant for structural studies

      • Consider cell-free expression systems for highly toxic constructs

The table below presents a systematic approach to troubleshooting expression issues:

IssueDiagnostic IndicatorsFirst-line SolutionAdvanced Solution
Inclusion bodiesInsoluble pellet in SDS-PAGELower temperature, reduce IPTGSwitch to solubility tag, co-express chaperones
DegradationMultiple bands below expected MWAdd protease inhibitorsUse protease-deficient strain
InactivityProtein present but no enzymatic activityTry different buffer conditionsCo-express with XseA
Low yieldFaint band on SDS-PAGEOptimize induction conditionsCodon optimization, change expression strain
ToxicityPoor growth post-inductionReduce inducer concentrationUse knockout strain, cell-free expression

By systematically addressing these challenges, researchers can significantly improve the likelihood of obtaining functional recombinant N. europaea XseB for subsequent structural and functional studies.

How can researchers reconcile contradictory findings on metal ion requirements for ExoVII activity when working with N. europaea XseB?

Contradictory findings regarding metal ion requirements for ExoVII activity, such as those observed between E. coli (metal-independent) and T. maritima (Mg²⁺-dependent) enzymes , present significant challenges for researchers working with N. europaea XseB. The following systematic approach can help reconcile such contradictions and establish definitive parameters for the N. europaea enzyme:

  • Comprehensive metal dependency screening:

    • Methodology: Perform nuclease activity assays under multiple conditions:

      • With various divalent cations (Mg²⁺, Mn²⁺, Ca²⁺, Zn²⁺, Co²⁺) at 1-10 mM concentrations

      • With metal chelators (EDTA, EGTA) at 1-20 mM

      • Under metal-free conditions using treated buffers and plasticware

    • Analysis: Quantify DNA degradation rates under each condition; construct activity profiles across metal concentrations

  • Protein sequence and structural analysis:

    • Methodology: Compare the amino acid sequences of N. europaea XseA and XseB with homologs from E. coli and T. maritima, focusing on:

      • Metal-binding motifs (e.g., DDE, DEDD, HxH motifs)

      • Conserved acidic residues in the catalytic domain

    • Analysis: Use structural prediction and molecular modeling to identify potential metal-binding sites

  • Site-directed mutagenesis:

    • Methodology: Based on sequence analysis, create point mutations in potential metal-binding residues in both XseA and XseB

    • Analysis: Test the metal dependency of each mutant to identify critical residues for metal coordination or metal-independent catalysis

  • Temperature and pH dependency correlation:

    • Methodology: Test metal requirements across a range of temperatures (10-40°C) and pH values (6.0-9.0)

    • Analysis: Determine if metal dependency varies with these conditions, which may explain some contradictions in previous studies

  • Substrate specificity assessment:

    • Methodology: Test metal requirements using different DNA substrates:

      • Various lengths of ssDNA (15-100 nucleotides)

      • Different DNA structures (5' overhangs, 3' overhangs, nicks)

      • RNA substrates

    • Analysis: Determine if metal requirements are substrate-specific

  • Kinetic analysis:

    • Methodology: Perform detailed enzyme kinetics (Km, Vmax) with and without metals

    • Analysis: Determine if metals affect substrate binding (Km) or catalytic rate (Vmax)

Based on the differing metal requirements observed between E. coli and T. maritima ExoVII , researchers should consider evolutionary and environmental factors that might influence metal dependency. N. europaea inhabits environments that may have different metal availabilities compared to E. coli or T. maritima habitats, potentially leading to adaptations in metal usage by its enzymes.

The table below outlines a decision matrix for determining the nature of metal involvement in N. europaea ExoVII:

Experimental ObservationInterpretationFollow-up Experiment
Activity with EDTA = Activity without EDTAMetal-independent mechanismTest structural role of metals using thermal stability assays
Activity with EDTA < Activity without EDTAMetal may have structural roleTest activity restoration by adding back specific metals
Activity only with specific metal(s)Metal is catalytically essentialPerform metal binding assays (ITC, MST) to confirm direct binding
Activity varies with metal:substrate ratioMetal mediates substrate interactionTest DNA binding with/without metals using EMSA
Temperature alters metal dependencyMetal role is condition-dependentDetermine thermodynamic parameters under different conditions

By systematically investigating these aspects, researchers can reconcile contradictory findings and establish a clear understanding of the metal ion requirements for N. europaea ExoVII, contributing valuable knowledge to the field of bacterial nucleases and DNA repair mechanisms.

How does the genetic organization of the xseB locus in N. europaea compare with other bacterial species, and what are the evolutionary implications?

The genetic organization of the xseB locus in N. europaea provides important insights into the evolution and functional conservation of Exodeoxyribonuclease VII across bacterial lineages. Through comparative genomic analysis, researchers can uncover important patterns that inform both fundamental understanding and experimental approaches.

  • Synteny analysis: In many proteobacteria, xseB is found in conserved gene neighborhoods, often near DNA repair and recombination genes. The preservation or disruption of this synteny in N. europaea provides clues about the evolutionary forces acting on this locus.

  • Operon structure: In some bacteria, xseA and xseB are arranged in an operon with coordinated expression. N. europaea may show variations in this arrangement, potentially reflecting adaptations to its specialized ecological niche as an ammonia oxidizer.

  • Regulatory elements: Identification of promoter regions and transcription factor binding sites upstream of xseB in N. europaea could reveal distinctive regulatory mechanisms compared to other bacteria.

  • Horizontal gene transfer assessment: Analyzing GC content, codon usage bias, and phylogenetic incongruence can determine if the xseB gene in N. europaea was acquired through horizontal gene transfer or has been maintained through vertical inheritance.

  • Selection pressure analysis: Calculating the ratio of non-synonymous to synonymous substitutions (dN/dS) in xseB across different bacterial lineages can identify regions under purifying or diversifying selection, providing insights into functional constraints.

Evolutionary implications of these comparisons include:

  • The conservation of the ExoVII complex architecture (1:4 ratio of XseA:XseB) across diverse bacteria suggests strong functional constraints on this quaternary structure .

  • Variations in metal ion requirements between bacterial species (e.g., E. coli vs. T. maritima) may reflect adaptations to different environmental conditions.

  • The presence or absence of compensatory DNA repair mechanisms in different bacteria may influence the evolutionary trajectory of xseB.

This evolutionary context is crucial for designing experiments that account for species-specific features of the N. europaea xseB gene and interpreting results in a broader biological framework.

What insights can comparative transcriptomics provide about the regulation of xseB expression under environmental stressors in N. europaea versus other bacteria?

Comparative transcriptomics offers powerful insights into the regulation of xseB expression under environmental stressors in N. europaea compared to other bacterial species. This approach can reveal both conserved and species-specific regulatory mechanisms that have evolved in response to different ecological niches.

When designing comparative transcriptomic studies focusing on xseB regulation, researchers should consider:

  • Selection of comparative species:

    • Include phylogenetically related ammonia-oxidizing bacteria (AOB) like Nitrosospira multiformis

    • Include model organisms with well-characterized DNA repair systems (E. coli, B. subtilis)

    • Include bacteria from similar environmental niches but different phylogenetic backgrounds

  • Environmental stressor selection:

    • Stressors relevant to N. europaea's ecology:

      • Varying ammonia concentrations (substrate-induced stress)

      • Chlorinated compounds (found in wastewater environments)

      • Oxygen limitation (common in biofilms and wastewater)

    • Universal stressors for cross-species comparison:

      • Oxidative stress (H₂O₂)

      • DNA-damaging agents (UV, mitomycin C)

      • Temperature shifts

  • Integrated analysis approaches:

    • Identify shared and species-specific transcriptional responses

    • Map regulons and potential transcription factor binding sites

    • Correlate expression patterns with known stress response pathways

    • Compare with metabolomic profiles to link transcriptional changes to physiological responses

Based on existing data from N. europaea under various stress conditions , several patterns might emerge from comparative transcriptomics:

  • Ammonia oxidation-specific regulation: In N. europaea, xseB expression may be coordinated with ammonia oxidation genes (amo, hao) under specific stressors, a pattern absent in non-AOB species.

  • Nitric oxide response network: Given the production of NO during ammonia oxidation, N. europaea might show distinctive co-regulation of xseB with nitrogen oxide metabolism genes (nirK, norB) compared to other bacteria .

  • Integration with energy metabolism: Since N. europaea is an obligate chemolithoautotroph, environmental stressors affecting energy production might show unique patterns of xseB regulation compared to heterotrophic bacteria.

  • Temporal expression dynamics: The kinetics of xseB expression in response to stressors may differ between N. europaea and other bacteria, reflecting different priorities in stress response allocation.

For experimental design, the table below outlines key comparisons and expected insights:

Comparison TypeExperimental ApproachExpected Insights
Cross-species under identical stressorsRNA-seq of N. europaea, E. coli, and other bacteria under standardized H₂O₂ exposureIdentify conserved vs. species-specific elements of oxidative stress response
Cross-condition for N. europaeaRNA-seq under chloroform, O₂ limitation, and ammonia variationMap condition-specific regulatory networks involving xseB
Temporal dynamicsTime-course RNA-seq after stressor applicationDetermine if xseB is part of early or late stress response
Regulatory mutantsTranscriptomics of N. europaea with mutations in key regulatorsIdentify regulatory factors controlling xseB expression

This comparative approach will provide a comprehensive understanding of how xseB regulation has evolved in N. europaea to meet the specific challenges of its ecological niche, while also revealing fundamental aspects of bacterial stress response systems conserved across diverse species.

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.