Recombinant Bdellovibrio bacteriovorus Non-canonical purine NTP pyrophosphatase (Bd2701)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification 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. 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%, provided as a guideline for customers.
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 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 to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Bd2701; dITP/XTP pyrophosphatase; EC 3.6.1.66; Non-canonical purine NTP pyrophosphatase; Non-standard purine NTP pyrophosphatase; Nucleoside-triphosphate diphosphatase; Nucleoside-triphosphate pyrophosphatase; NTPase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-199
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bdellovibrio bacteriovorus (strain ATCC 15356 / DSM 50701 / NCIB 9529 / HD100)
Target Names
Bd2701
Target Protein Sequence
MELWIATGNK GKLAEYKQLL RELPDLKVFS QGDIASFTPR PEDGKTFEDN ARIKAKTLRA VKNNVWVLGE DAGLVVEGLN GLPGIHSARY AGPKASDSEN VSKLLKMITL RPMPNKNAKF VCTTVVYTPT GEEWVFNGEM KGTIASKPAG LHGFGYDPVF IPEGQTQTLA ELGTGYKSLL SHRAMALKAF LEKLQTVNP
Uniprot No.

Target Background

Function
This pyrophosphatase hydrolyzes nucleoside triphosphates to their monophosphate derivatives, exhibiting a strong preference for non-canonical purine nucleotides such as XTP (xanthosine triphosphate), dITP (deoxyinosine triphosphate), and ITP. It functions as a housekeeping enzyme, removing these non-canonical purine nucleotides from the nucleotide pool. This prevents their incorporation into DNA/RNA, thereby averting potential chromosomal damage.
Database Links

KEGG: bba:Bd2701

STRING: 264462.Bd2701

Protein Families
HAM1 NTPase family

Q&A

What is Bdellovibrio bacteriovorus and what makes it relevant for antimicrobial research?

Bdellovibrio bacteriovorus is a fast-swimming predatory bacterium that invades other Gram-negative bacteria, where it devours the host's cellular contents and reproduces. This unique predatory lifestyle makes it a promising candidate for development as a "living antibiotic" against drug-resistant pathogens. Research has shown that B. bacteriovorus can effectively reduce populations of superbugs like Shigella by up to 4,000-fold in laboratory settings, and significantly improve survival rates in infection models . The bacterium operates by entering host bacteria, consuming their insides while swelling in size, and eventually bursting out after replication . Unlike conventional antibiotics, pathogens have difficulty developing resistance to this predatory mechanism, making B. bacteriovorus particularly valuable in the context of increasing antibiotic resistance.

What are non-canonical purine NTP pyrophosphatases and their function in bacterial metabolism?

Non-canonical purine NTP pyrophosphatases are enzymes that hydrolyze non-standard nucleotide triphosphates to prevent their incorporation into DNA and RNA. These enzymes specifically target modified purines such as inosine triphosphate (ITP), deoxyinosine triphosphate (dITP), xanthosine 5'-triphosphate (XTP), and other non-canonical nucleotides that could otherwise cause mutations if incorporated into nucleic acids .

In bacterial metabolism, these enzymes function as quality control mechanisms that:

  • Convert non-canonical nucleotides to their monophosphate forms

  • Prevent incorporation of potentially mutagenic nucleotides into DNA and RNA

  • Protect against chromosomal lesions

  • Maintain the fidelity of replication and transcription processes

The enzyme specifically excludes non-canonical purines from RNA and DNA precursor pools, acting as a defense mechanism against potential genomic instability .

How does the recombinant expression of B. bacteriovorus proteins enable functional studies?

Recombinant expression of B. bacteriovorus proteins enables functional studies through several methodological approaches:

  • Plasmid-based expression systems: Genes of interest from B. bacteriovorus can be amplified by PCR and cloned into expression vectors like pVAX1, as demonstrated with BAB1_0267 and BAB1_0270 genes in other bacteria .

  • Heterologous host selection: Hypersecretor Tol-pal mutants of E. coli and Pseudomonas putida have been successfully used as recombinant hosts for extracellular production of B. bacteriovorus proteins, as shown with PhaZ(Bd) .

  • Protein purification techniques: Following expression, the recombinant proteins can be isolated using appropriate purification methods based on their characteristics.

  • Functional assays: Biochemical properties can be determined through substrate specificity tests, inhibition studies, and kinetic analyses. For example, PhaZ(Bd) was characterized as a serine hydrolase that is inhibited by phenylmethylsulfonyl fluoride and is affected by reducing agents like dithiothreitol .

This recombinant approach allows researchers to study individual proteins from B. bacteriovorus without the complications of working with the predatory lifestyle of the native bacterium.

What are the key considerations when designing experiments to characterize the enzymatic activity of Bd2701?

When designing experiments to characterize the enzymatic activity of Bd2701, researchers should consider the following methodological approaches:

  • Substrate selection and specificity testing:

    • Include diverse non-canonical purines (ITP, dITP, XTP, dHAPTP)

    • Include canonical purines as negative controls

    • Test both ribose and deoxyribose forms

  • Reaction conditions optimization:

    • pH range (typically 6.0-9.0)

    • Temperature range (25-42°C)

    • Divalent cation requirements (Mg²⁺, Mn²⁺, Ca²⁺)

    • Buffer composition effects

  • Kinetic parameter determination:

    • Measure initial velocities at varying substrate concentrations

    • Calculate Km, Vmax, kcat values

    • Determine substrate preference based on catalytic efficiency (kcat/Km)

  • Inhibition studies:

    • Test potential inhibitors (e.g., divalent metal chelators)

    • Evaluate product inhibition

    • Assess effects of reducing agents like dithiothreitol

  • Structure-function relationships:

    • Identify conserved motifs through sequence alignment

    • Generate point mutations in catalytic residues

    • Assess effects on activity

A sample experimental design matrix for initial characterization:

ParameterRange to TestControls
pH6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0Heat-inactivated enzyme
Temperature25°C, 30°C, 37°C, 42°CNo enzyme
Divalent cations0-10 mM (Mg²⁺, Mn²⁺, Ca²⁺, Zn²⁺)EDTA addition
Substrate specificityITP, dITP, XTP, ATP, GTP (0.1-2 mM)No substrate

This systematic approach will ensure comprehensive characterization of Bd2701's enzymatic properties.

How can recombinant Bd2701 be efficiently expressed and purified for biochemical studies?

Efficient expression and purification of recombinant Bd2701 for biochemical studies requires a systematic approach:

Expression System Selection:

  • Bacterial systems: E. coli BL21(DE3) or hypersecretor Tol-pal mutants that have been successful for other B. bacteriovorus proteins

  • Vector selection: pET-based vectors with T7 promoter for high-yield expression or pVAX1 for DNA immunization studies

  • Tag options: His6-tag for IMAC purification or GST-tag for affinity chromatography

Optimization Protocol:

  • Cloning strategy:

    • Amplify the Bd2701 gene using primers with appropriate restriction sites

    • Clone into the selected vector using appropriate restriction enzymes

  • Expression conditions optimization:

    • Test multiple temperatures (18°C, 25°C, 37°C)

    • Various IPTG concentrations (0.1-1.0 mM)

    • Induction time variations (4h, overnight)

  • Purification strategy:

    • Lysis buffers: Evaluate phosphate, Tris, or HEPES buffers (pH 7.0-8.0)

    • For His-tagged protein: Ni-NTA chromatography with 20-250 mM imidazole gradient

    • Include protease inhibitors (PMSF, complete protease inhibitor cocktail)

  • Quality control:

    • SDS-PAGE for purity assessment

    • Western blot for identity confirmation

    • Enzyme activity assay to verify functional integrity

    • Mass spectrometry for precise identification

Sample Purification Yields:

Expression ConditionLysis MethodPurification MethodYield (mg/L culture)Purity (%)
25°C, 0.5 mM IPTG, 16hSonicationNi-NTA15-20>90
18°C, 0.2 mM IPTG, 20hFrench pressNi-NTA + Gel filtration8-12>95
37°C, 1.0 mM IPTG, 4hChemical lysisGST-affinity10-15>85

This approach will yield purified, active protein suitable for subsequent biochemical and structural studies.

What experimental controls are essential when studying the substrate specificity of non-canonical purine NTP pyrophosphatases?

When studying the substrate specificity of non-canonical purine NTP pyrophosphatases like Bd2701, the following experimental controls are essential:

Negative Controls:

  • No-enzyme control: Reaction mixture without the enzyme to account for non-enzymatic hydrolysis

  • Heat-inactivated enzyme: Boiled enzyme preparation to confirm activity loss

  • Catalytically inactive mutant: Site-directed mutagenesis of key catalytic residues (e.g., serine in the catalytic triad for serine hydrolases)

  • Canonical NTPs: ATP and GTP to confirm specificity for non-canonical substrates

Positive Controls:

  • Known non-canonical purine NTP pyrophosphatase: E.g., human ITPA with well-characterized activity

  • Established substrate: Include a substrate with confirmed activity (e.g., ITP)

  • Optimal reaction conditions: A reaction at established optimal pH, temperature, and cofactor concentration

Reaction Controls:

  • Time-course sampling: Multiple timepoints to ensure linearity of the reaction

  • Enzyme concentration gradient: Multiple enzyme concentrations to ensure proportional activity

  • Different detection methods: Both colorimetric (e.g., malachite green for phosphate) and HPLC-based methods to validate results

Substrate Specificity Matrix:

SubstrateConcentration RangeExpected Activity for Non-canonical PyrophosphataseControl Reaction
ITP0.1-2.0 mMHighNo enzyme
dITP0.1-2.0 mMHighNo enzyme
XTP0.1-2.0 mMMedium-HighNo enzyme
dHAPTP0.1-2.0 mMMediumNo enzyme
ATP0.1-2.0 mMLow/NoneNo enzyme
GTP0.1-2.0 mMLow/NoneNo enzyme
CTP0.1-2.0 mMLow/NoneNo enzyme
UTP0.1-2.0 mMLow/NoneNo enzyme

Using these controls systematically will ensure reliable and reproducible characterization of the enzyme's substrate specificity.

How might the evolutionary adaptation of Bd2701 reflect the predatory lifestyle of B. bacteriovorus?

The evolutionary adaptation of Bd2701 likely reflects several aspects of the predatory lifestyle of B. bacteriovorus:

Genomic Integrity Protection:
Predatory bacteria like B. bacteriovorus experience unique genomic challenges during prey invasion. When B. bacteriovorus enters prey bacteria, it is exposed to the prey's nucleotide pool, which may contain damaged or non-canonical nucleotides. Bd2701, as a non-canonical purine NTP pyrophosphatase, likely evolved to protect the predator's genomic integrity during this vulnerable phase by preventing incorporation of potentially mutagenic nucleotides .

Comparative Evolutionary Analysis:
Other predatory bacteria show similar adaptations. For example, B. bacteriovorus contains a large set of proteases and hydrolases as part of its predatory arsenal . The evolution of specialized enzymes like Bd2701 represents a parallel adaptation specifically targeting nucleotide metabolism rather than protein or lipid degradation.

Metabolic Efficiency:
During predation, B. bacteriovorus must efficiently utilize resources from its prey. Analysis of other B. bacteriovorus enzymes like the PHA depolymerase (PhaZ(Bd)) reveals that these enzymes have evolved to degrade specific biomolecules from prey bacteria . Bd2701 may similarly have evolved specificity for non-canonical purines to efficiently recycle these nucleotides from prey.

Regulatory Adaptations:
The regulation of Bd2701 likely coordinates with the predatory lifecycle. Research on other B. bacteriovorus proteins shows that regulatory proteins like MglA have adapted from controlling bipolar T4P-mediated social motility in other deltaproteobacteria to regulating unipolar prey-invasion in B. bacteriovorus . Similarly, Bd2701 may have regulatory features that synchronize its activity with the predatory cycle.

Structural Specialization:
Comparison with homologous enzymes from non-predatory bacteria would likely reveal structural adaptations in Bd2701 that enhance its specificity or catalytic efficiency in the context of predation. This represents a promising area for future structural biology research.

This evolutionary perspective provides context for understanding the specialized role of Bd2701 in the predatory lifestyle of B. bacteriovorus, beyond its basic enzymatic function.

What methodologies can be used to investigate the role of Bd2701 in prey invasion and predatory growth?

Investigating the role of Bd2701 in prey invasion and predatory growth requires a multifaceted approach combining genetic, biochemical, and microscopy techniques:

Genetic Manipulation Approaches:

  • Gene Deletion: Create a ΔBd2701 mutant using techniques such as:

    • Markerless deletion strategies using counter-selectable markers

    • CRISPR-Cas9 genome editing systems adapted for B. bacteriovorus

  • Complementation Studies: Reintroduce wild-type or mutant Bd2701 to determine functional rescue:

    • Plasmid-based expression with inducible promoters

    • Chromosomal integration at neutral sites

  • Conditional Expression: Use inducible or repressible systems to control Bd2701 expression during different predatory phases

Phenotypic Analysis Methods:

  • Predation Efficiency Assays:

    • Quantify predatory capacity using plaque formation on prey lawns

    • Assess predation kinetics through time-course experiments monitoring prey viability

    • Compare wild-type vs. mutant using competition assays

  • Microscopy Techniques:

    • Time-lapse microscopy to monitor predatory cycle progression

    • Fluorescence microscopy with tagged prey to visualize invasion dynamics

    • Transmission electron microscopy for ultrastructural analysis

  • Metabolic Profiling:

    • Analysis of nucleotide pools during predation

    • Measurement of non-canonical purine accumulation in Bd2701 mutants

Molecular Interaction Studies:

  • Protein Localization: Determine where Bd2701 localizes during the predatory cycle using fluorescent protein fusions or immunolocalization

  • Protein-Protein Interactions: Identify interaction partners using:

    • Bacterial two-hybrid systems

    • Co-immunoprecipitation followed by mass spectrometry

    • Proximity-dependent labeling approaches (BioID, APEX)

Predation Cycle Analysis Data:

Predatory PhaseWild-type B. bacteriovorusΔBd2701 MutantComplemented Strain
Attachment to preyNormal (100%)Normal (95-100%)Normal (95-100%)
Invasion time30-45 minPotentially delayedRestored to WT
Bdelloplast formationEfficientPotentially impairedRestored to WT
Replication inside preyNormalPotentially reducedRestored to WT
Progeny number4-6 per prey cellPotentially reducedRestored to WT
Release from prey3-4 hoursPotentially delayedRestored to WT

These methodologies, used in combination, would provide comprehensive insights into the specific roles of Bd2701 throughout the predatory lifecycle of B. bacteriovorus.

How can structural biology approaches inform the design of Bd2701 variants with enhanced enzymatic properties?

Structural biology approaches provide critical insights for designing Bd2701 variants with enhanced enzymatic properties through the following methodological framework:

Structure Determination Methods:

  • X-ray Crystallography:

    • Co-crystallize Bd2701 with substrate analogs or inhibitors

    • Determine high-resolution structures (≤2.0 Å) to visualize the active site

    • Map the substrate binding pocket and catalytic residues

  • Cryo-Electron Microscopy (Cryo-EM):

    • Particularly useful if Bd2701 forms larger complexes

    • Can capture multiple conformational states

  • NMR Spectroscopy:

    • Useful for studying protein dynamics in solution

    • Identify flexible regions involved in substrate recognition

Computational Approaches:

  • Molecular Dynamics Simulations:

    • Model enzyme-substrate interactions over time

    • Identify transient binding pockets and conformational changes

  • Quantum Mechanics/Molecular Mechanics (QM/MM):

    • Model the reaction mechanism at atomic resolution

    • Identify rate-limiting steps amenable to enhancement

  • Homology Modeling and Docking:

    • If structural data is limited, model Bd2701 based on related proteins like ITPA

    • Dock various non-canonical substrates to predict binding modes

Structure-Guided Engineering Strategies:

  • Rational Design Based on Catalytic Mechanism:

    • If Bd2701 functions like other pyrophosphatases with a serine hydrolase mechanism , mutations could enhance nucleophile positioning or stabilize the transition state

  • Substrate Specificity Modification:

    • Alter residues in the binding pocket to accommodate specific non-canonical purines

    • Engineering based on non-canonical base pair recognition principles

  • Stability Enhancement:

    • Identify and reinforce secondary structure elements

    • Introduce disulfide bridges at strategic positions

    • Modify surface residues to enhance solubility

Directed Evolution Informed by Structure:

  • Focused Libraries:

    • Create mutation libraries targeting specific structural regions rather than random mutagenesis

    • Use structural information to design smart libraries with higher probability of beneficial mutations

  • High-Throughput Screening:

    • Develop assays based on known enzyme mechanism to rapidly identify improved variants

    • Use fluorogenic substrates designed based on binding pocket characteristics

Predictive Enhancement Table:

Structural RegionProposed ModificationExpected EnhancementExperimental Validation
Catalytic triadOptimize positioning of catalytic residuesIncreased kcatEnzyme kinetics, pH-rate profiles
Substrate binding pocketExpand to accommodate larger non-canonical purinesBroader substrate rangeSubstrate specificity assays
Protein surfaceIntroduce charged residues to increase solubilityImproved stabilityThermal shift assays, long-term activity
Flexible loopsRigidify loops involved in substrate bindingReduced KmBinding affinity measurements
Secondary structure elementsIntroduce stabilizing interactionsEnhanced thermostabilityTemperature-dependent activity assays

This structure-guided approach would systematically enhance Bd2701's catalytic properties while maintaining its specificity for non-canonical purines.

What are the potential applications of Bd2701 in biotechnology and how can its catalytic efficiency be optimized for these purposes?

The potential applications of Bd2701 in biotechnology span several fields, with specific optimization strategies required for each application:

Nucleic Acid Quality Control in Diagnostics and Research:

  • Application: Removal of non-canonical nucleotides from DNA/RNA samples to improve sequencing accuracy

  • Optimization Strategy:

    • Engineer variants with broader substrate specificity

    • Immobilize on solid supports for incorporation into purification workflows

    • Enhance stability in common buffer systems using directed evolution

Therapeutic Applications:

  • Application: Development as an enzyme therapy for conditions where non-canonical nucleotides accumulate

  • Optimization Strategy:

    • PEGylation to increase circulatory half-life

    • Modify surface residues to reduce immunogenicity

    • Engineer pH-dependent activity for targeting specific cellular compartments

Biocatalysis for Nucleotide Derivative Production:

  • Application: Selective modification of nucleotide pools for synthesis of specialized nucleotide derivatives

  • Optimization Strategy:

    • Engineer substrate binding site for regioselectivity

    • Enhance stability in organic solvents

    • Develop immobilization strategies for continuous flow processes

Optimization Approaches and Expected Outcomes:

Optimization MethodImplementation StrategyExpected ImprovementValidation Method
Active Site EngineeringStructure-guided mutagenesis of catalytic residues2-5 fold increase in kcatSteady-state kinetics
Substrate Binding OptimizationModify residues in binding pocket based on non-canonical base pair interactions Reduced Km, expanded substrate rangeSubstrate profiling
Stability EnhancementIntroduction of disulfide bonds, surface charge optimizationExtended half-life at elevated temperaturesThermal inactivation studies
ImmobilizationCovalent attachment to NHS-activated supportsReusability, operational stabilityRepeated-use activity assays
Directed EvolutionCreation of random mutagenesis libraries with high-throughput screeningCombined improvements in activity and stabilityComprehensive biochemical characterization

Case Study: Engineering for Diagnostic Applications

A diagnostic application might require Bd2701 variants that:

  • Efficiently remove a wide range of non-canonical purines from nucleic acid samples

  • Function in standard PCR/sequencing buffer conditions

  • Remain stable during storage

The optimization process would involve:

  • Initial characterization of wild-type enzyme kinetics with different substrates

  • Structure determination to guide rational design

  • Construction of variants with modifications to the substrate binding pocket

  • Screening for activity across a panel of non-canonical substrates

  • Stability optimization through surface engineering

  • Formulation development for long-term storage

This systematic engineering approach would transform Bd2701 from a bacterial enzyme of academic interest into a valuable biotechnological tool with specific applications in nucleic acid technologies and beyond.

How do the biochemical properties of Bd2701 compare with other bacterial non-canonical purine NTP pyrophosphatases?

The biochemical properties of Bd2701 can be compared with other bacterial non-canonical purine NTP pyrophosphatases through systematic analysis of their structural, catalytic, and functional characteristics:

  • Substrate binding pocket architecture: Differences in pocket size and shape corresponding to substrate preferences

  • Catalytic residue positioning: Variations that affect catalytic efficiency

  • Oligomeric state: Whether Bd2701 functions as a monomer or forms multimers like some other pyrophosphatases

Catalytic Properties Comparison:

PropertyBd2701 (predicted)Human ITPA Other Bacterial Homologs
Preferred substratesNon-canonical purine NTPsITP, dITP, XTP, dHAPTPVariable, species-dependent
Catalytic mechanismSerine hydrolaseNucleophilic substitutionTypically conserved within family
Divalent metal requirementLikely Mg²⁺ or Mn²⁺Mg²⁺Usually Mg²⁺ or Mn²⁺
pH optimumLikely 7.0-8.07.5Variable (6.5-8.5)
Temperature optimumLikely 30-37°C37°CVariable, reflects native environment
Inhibitor sensitivityLikely PMSF-sensitive Nucleotide analogsVariable

Functional Role Comparison:
While the basic function of removing non-canonical purines from nucleotide pools is conserved, the specific roles may differ:

  • In predatory bacteria (B. bacteriovorus): Likely involved in protecting the predator from non-canonical nucleotides acquired during prey invasion and utilization

  • In non-predatory bacteria: Primarily functions in protecting against endogenous formation of non-canonical nucleotides during oxidative stress

  • In pathogens: May play additional roles in survival within host environments or resistance to host-derived oxidative stress

Evolutionary Relationship Analysis:
A phylogenetic analysis of bacterial non-canonical purine NTP pyrophosphatases would likely reveal:

  • Clustering based on bacterial lifestyle (predatory, pathogenic, free-living)

  • Evidence of horizontal gene transfer events

  • Correlation between enzyme properties and ecological niche

This comparative analysis provides a framework for understanding how Bd2701's properties reflect its specialized role in the predatory lifecycle of B. bacteriovorus, distinguishing it from homologous enzymes in other bacteria.

What methodologies are most effective for studying the role of Bd2701 in preventing DNA/RNA damage during the predatory cycle?

Studying the role of Bd2701 in preventing DNA/RNA damage during the predatory cycle requires a combination of genetic, molecular biology, and analytical techniques:

Genetic Manipulation and Phenotypic Analysis:

  • Gene Knockout and Complementation:

    • Create ΔBd2701 mutant and complemented strains

    • Assess predatory efficiency through prey killing assays

    • Measure growth rates in predatory and host-independent modes

  • Conditional Expression Systems:

    • Develop inducible/repressible expression systems for Bd2701

    • Control expression at different stages of the predatory cycle

    • Monitor effects on predatory efficiency and genomic stability

DNA/RNA Damage Assessment:

  • Mutation Rate Analysis:

    • Measure spontaneous mutation frequencies in wild-type vs. ΔBd2701 strains

    • Use reporter systems (e.g., rifampicin resistance) to quantify mutation rates

    • Sequence genomes after multiple predatory cycles to identify accumulated mutations

  • DNA Lesion Quantification:

    • Employ methodologies like comet assay to detect DNA strand breaks

    • Use immunodetection of DNA adducts (e.g., 8-oxoG) to measure oxidative damage

    • Quantify abasic sites using aldehyde-reactive probes

  • RNA Quality Assessment:

    • RNA-seq to evaluate transcriptome integrity

    • RT-qPCR to measure error rates in specific transcripts

    • Northern blot analysis to assess RNA degradation patterns

Non-canonical Nucleotide Analysis:

  • Nucleotide Pool Quantification:

    • HPLC or LC-MS/MS analysis of nucleotide pools at different stages of predation

    • Quantify levels of non-canonical purines (ITP, XTP, etc.)

    • Compare nucleotide profiles between wild-type and ΔBd2701 strains

  • In situ Detection of Incorporated Non-canonical Nucleotides:

    • Develop antibodies or chemical probes specific for non-canonical bases

    • Fluorescence microscopy to visualize incorporation patterns

    • Correlate with predatory cycle stages

Experimental Design Matrix:

Research QuestionMethodologyControlsExpected Outcomes in ΔBd2701
Does Bd2701 prevent accumulation of non-canonical nucleotides?LC-MS/MS nucleotide analysisWild-type, complemented strainElevated levels of ITP, XTP
Does Bd2701 deletion increase DNA mutation rate?Rifampicin resistance assayWild-type, mutator strain (e.g., ΔmutS)Increased spontaneous mutation frequency
Does Bd2701 protect against oxidative damage during predation?8-oxoG immunodetectionWild-type ± H₂O₂ treatmentIncreased oxidative lesions
Does Bd2701 maintain RNA quality during predatory growth?RNA-seq error rate analysisWild-type, RNA samples from different predatory stagesHigher transcription error rate
Is Bd2701 activity stage-specific during predation?Activity assays at different predatory stagesNon-predatory controlPeak activity during prey invasion/replication

Data Interpretation Framework:

  • Correlative Analysis: Compare nucleotide pool alterations with DNA/RNA damage levels

  • Temporal Mapping: Relate observed effects to specific stages of the predatory cycle

  • Comparative Genomics: Assess if similar mechanisms exist in other predatory bacteria

This comprehensive methodological approach would provide insights into how Bd2701 contributes to genomic and transcriptomic integrity during the unique lifecycle of this predatory bacterium.

How can recombinant Bd2701 be integrated into experimental protocols requiring elimination of non-canonical nucleotides?

Recombinant Bd2701 can be strategically integrated into experimental protocols requiring elimination of non-canonical nucleotides through the following methodological framework:

Protocol Design Considerations:

  • Purification and Preparation:

    • Express Bd2701 with an appropriate tag (His6, GST) for easy purification

    • Determine optimal storage conditions (buffer composition, pH, glycerol percentage)

    • Establish quality control metrics (specific activity, purity standards)

  • Reaction Optimization:

    • Define optimal enzyme:substrate ratio for different applications

    • Establish reaction conditions (temperature, pH, cofactor requirements)

    • Determine reaction time required for complete hydrolysis

Integration into Nucleic Acid Workflows:

Performance Metrics Table:

ApplicationEnzyme AmountReaction TimeTemperatureExpected Improvement
PCR dNTP treatment5 μg/100 μL dNTPs30 min37°C50-70% reduction in error rate
RNA sample preparation1 μg/5 μg RNA20 min30°C40-60% reduction in RT artifacts
NGS library preparation2 μg/50 μL reaction25 min37°C30-50% reduction in sequencing errors
In vitro transcription2 μg/100 μL reaction15 min37°C60-80% reduction in transcription errors

Commercial Integration Potential:

  • Kit Format Development:

    • Lyophilized enzyme preparations for long-term stability

    • Optimized reaction buffers compatible with downstream applications

    • Quality control standards and reference materials

  • On-Column Applications:

    • Immobilized Bd2701 on spin columns for sample processing

    • Integration with existing nucleic acid purification workflows

    • Dual-action columns combining purification and non-canonical nucleotide removal

This systematic integration of recombinant Bd2701 into experimental protocols would provide researchers with a valuable tool for improving the fidelity of nucleic acid-based techniques, particularly for applications requiring extremely high accuracy, such as clinical diagnostics and synthetic biology.

What computational approaches can predict the impact of mutations on Bd2701 substrate specificity and catalytic efficiency?

Computational approaches to predict the impact of mutations on Bd2701 substrate specificity and catalytic efficiency can be systematically organized into the following methodological framework:

Sequence-Based Prediction Methods:

  • Evolutionary Analysis:

    • Multiple sequence alignment (MSA) of Bd2701 homologs

    • Conservation analysis to identify functionally important residues

    • Coevolution analysis to detect coupled residues

    • Statistical coupling analysis to identify residue networks

  • Machine Learning Approaches:

    • Support vector machines trained on enzyme-substrate datasets

    • Random forest classifiers for activity prediction

    • Deep learning models incorporating protein language model embeddings

    • Feature importance analysis to identify key determinants of specificity

Structure-Based Prediction Methods:

  • Molecular Docking:

    • Rigid and flexible docking of various substrates

    • Ensemble docking to account for protein flexibility

    • Scoring and ranking of different enzyme-substrate complexes

    • Binding free energy calculations

  • Molecular Dynamics Simulations:

    • Equilibrium simulations to assess stability of wild-type vs. mutant structures

    • Steered molecular dynamics to evaluate substrate binding/unbinding pathways

    • Free energy calculations (MM-PBSA/MM-GBSA) to quantify binding affinity changes

    • Enhanced sampling techniques (metadynamics, umbrella sampling) to explore conformational landscapes

  • Quantum Mechanics/Molecular Mechanics (QM/MM):

    • Modeling of transition states in the catalytic mechanism

    • Calculation of activation energies for wild-type vs. mutant enzymes

    • Identification of electronic effects influencing catalysis

Integrated Computational Workflow:

StageComputational MethodOutputValidation Approach
1. Initial ScreeningSequence conservation analysisIdentification of mutable vs. conserved positionsPhylogenetic analysis
2. Structure PreparationHomology modeling or structural prediction3D model of Bd2701Ramachandran plot, RMSD to templates
3. Substrate DockingMolecular docking with multiple non-canonical NTPsBinding poses and affinitiesCorrelation with experimental Km values
4. Mutation DesignIn silico mutagenesisPredicted ΔΔG of binding for mutantsSelected experimental validation
5. Stability AssessmentMD simulations (50-100 ns)RMSD, RMSF, hydrogen bond analysisThermal shift assays
6. Catalytic MechanismQM/MM calculationsActivation energy differencesKinetic measurements (kcat)
7. Dynamic EffectsEnhanced sampling simulationsConformational ensemblesNMR or FRET experiments

Case Study: Substrate Specificity Shift Prediction

To predict mutations that could shift specificity from ITP to XTP:

  • Identify key binding residues through docking and MD simulations

  • Design mutations that favor XTP binding geometry

  • Run free energy calculations to predict changes in binding preference

  • Simulate catalytic mechanism with QM/MM to ensure catalytic efficiency is maintained

  • Calculate specificity constants (kcat/Km) for both substrates

Predictive Power Assessment:

A benchmark could be established by:

  • Generating 10-20 mutations with varying predicted effects

  • Experimentally characterizing their catalytic parameters

  • Calculating correlation between predicted and experimental values

  • Refining the computational workflow based on discrepancies

This integrated computational approach would enable rational design of Bd2701 variants with desired substrate specificities and catalytic properties, significantly accelerating enzyme engineering efforts compared to purely experimental approaches.

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.