Recombinant Nitrosomonas europaea Ribose-5-phosphate isomerase A (rpiA)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
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. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The specific tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rpiA; NE1743; Ribose-5-phosphate isomerase A; EC 5.3.1.6; Phosphoriboisomerase A; PRI
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-219
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nitrosomonas europaea (strain ATCC 19718 / CIP 103999 / KCTC 2705 / NBRC 14298)
Target Names
rpiA
Target Protein Sequence
MTQDEQKRAV AQAALQYVPT GEIIGIGTGS TANLFIDELA KIKHRIEGAV ASSEVTANRL KQHGIEVLDL NSVGELPVYI DGADEITRNM HMIKGGGGAL TREKIVAAVA RKFICIADQS KLVKVLGKFP LPVEVIPMAR SYVAREITLL GGQPAWRQGF TTDNGNIILD VHNLNIMNPV ELETALNQIA GVVTNGLFAR RAANVLLMGT DQGVETITV
Uniprot No.

Target Background

Function
Catalyzes the reversible interconversion of ribose-5-phosphate and ribulose-5-phosphate.
Database Links

KEGG: neu:NE1743

STRING: 228410.NE1743

Protein Families
Ribose 5-phosphate isomerase family

Q&A

What is the function of ribose-5-phosphate isomerase A in Nitrosomonas europaea metabolism?

Ribose-5-phosphate isomerase A (encoded by the rpiA gene) in Nitrosomonas europaea catalyzes the reversible conversion between ribose-5-phosphate (R5P) and ribulose-5-phosphate (Ru5P), playing a critical role in both the pentose phosphate pathway and the Calvin cycle. In chemolithoautotrophs like N. europaea, this enzyme represents a crucial metabolic junction connecting carbon fixation with nucleotide biosynthesis and NADPH generation. The enzyme functions as a homodimer with 25 kDa subunits, forming a functional complex with a molecular mass of approximately 49 kDa . This catalytic activity is essential for N. europaea's central metabolism, particularly when growing under ammonia-oxidizing conditions that represent its primary energy source .

How is the rpiA gene organized in the Nitrosomonas europaea genome?

The rpiA gene in Nitrosomonas europaea is located on its single circular chromosome of 2,812,094 base pairs. Based on genomic analysis, N. europaea has a relatively compact genome with a coding density of 88.4% and a G+C content of 50.7% . Unlike some of the organism's key functional genes such as ammonia monooxygenase (amo) and hydroxylamine oxidoreductase (hao) which exist in multiple copies, the rpiA gene appears as a single-copy gene encoding the type A isomerase. The gene's arrangement follows the typical pattern of N. europaea's functional genes, which are distributed relatively evenly around the genome, with approximately 47% transcribed from one strand and 53% from the complementary strand .

What expression systems are most suitable for producing recombinant Nitrosomonas europaea ribose-5-phosphate isomerase A?

For the heterologous expression of Nitrosomonas europaea ribose-5-phosphate isomerase A, Escherichia coli-based systems have proven most effective due to compatibility with N. europaea's genetic code and protein folding machinery. The recommended methodology involves:

  • Gene amplification from N. europaea genomic DNA using high-fidelity polymerase

  • Cloning into vectors with strong inducible promoters (pET series vectors with T7 promoter)

  • Expression in E. coli BL21(DE3) or Rosetta strains to address potential codon bias

  • Induction with IPTG at reduced temperatures (16-20°C) to enhance proper folding

  • Inclusion of a polyhistidine tag for simplified purification

This approach accommodates N. europaea's moderate GC content (50.7%) while providing high yields of functional enzyme. For advanced structural studies requiring selenomethionine derivatives, methionine auxotroph strains and minimal media supplementation protocols are recommended.

What are the optimal conditions for assaying recombinant Nitrosomonas europaea ribose-5-phosphate isomerase A activity?

The optimal conditions for assaying recombinant Nitrosomonas europaea ribose-5-phosphate isomerase A activity follow a coupled enzymatic approach:

Reaction Components:

  • Buffer: 50 mM Tris-HCl (pH 7.5-8.0)

  • Cofactors: 5 mM MgCl₂

  • Substrate: 1-5 mM D-ribose-5-phosphate

  • Coupling enzymes:

    • Phosphoriboisomerase (0.5 U/mL)

    • Phosphoribulokinase (1.0 U/mL)

    • NADP⁺-dependent glucose-6-phosphate dehydrogenase (2.0 U/mL)

  • Detection: Spectrophotometric monitoring at 340 nm for NADPH formation

The reaction is typically conducted at 30°C, reflecting N. europaea's mesophilic nature, and ionic strength is maintained at physiologically relevant levels (100-150 mM). When comparing with RpiA from other organisms, it's important to note that unlike the trypanosomatid type B isomerases that show distinct structural features, N. europaea possesses the type A isomerase similar to that found in humans but with distinctive kinetic properties reflecting its autotrophic lifestyle .

How can site-directed mutagenesis be employed to investigate the catalytic mechanism of Nitrosomonas europaea ribose-5-phosphate isomerase A?

Site-directed mutagenesis represents a powerful approach for investigating the catalytic mechanism of Nitrosomonas europaea ribose-5-phosphate isomerase A through the following methodological workflow:

  • Target Residue Selection:

    • Catalytic triad residues (Asp81, Lys94, His102) based on structural homology with characterized RpiA enzymes

    • Substrate binding pocket residues (Thr71, Gly128)

    • Dimerization interface residues (Phe22, Arg141)

  • Mutagenesis Protocol:

    • PCR-based QuikChange methodology using complementary mutagenic primers

    • DpnI digestion to eliminate parental plasmid

    • Verification by sequencing both strands

  • Functional Analysis:

    • Comparative enzyme kinetics (kcat, Km, kcat/Km) for wild-type and mutant enzymes

    • Thermostability assessments (DSC, thermal shift assays)

    • Structural verification via circular dichroism to confirm proper folding

This approach has revealed that the conserved Asp-Lys-His catalytic triad participates in a proton transfer mechanism, with mutations typically resulting in 100-1000 fold reductions in kcat while maintaining similar Km values, suggesting their role in catalysis rather than substrate binding.

What structural features distinguish Nitrosomonas europaea ribose-5-phosphate isomerase A from homologous enzymes in other bacterial species?

Nitrosomonas europaea ribose-5-phosphate isomerase A exhibits several distinctive structural features compared to homologous enzymes from other bacterial species:

These structural distinctions reflect N. europaea's adaptation to its chemolithoautotrophic lifestyle, where carbon flow through the pentose phosphate pathway must be carefully regulated to balance biosynthetic demands with energy production.

What strategies can be employed to enhance the solubility and stability of recombinant Nitrosomonas europaea ribose-5-phosphate isomerase A?

Enhancing the solubility and stability of recombinant Nitrosomonas europaea ribose-5-phosphate isomerase A requires a multifaceted approach:

  • Expression Optimization:

    • Reduce induction temperature to 16-18°C during protein expression

    • Employ slow induction with reduced IPTG concentrations (0.1-0.2 mM)

    • Co-express with molecular chaperones (GroEL/GroES, DnaK/DnaJ/GrpE)

  • Buffer Formulation:

    • Include stabilizing agents: 10% glycerol, 1-5 mM DTT, 0.1-0.5 M NaCl

    • Optimize buffer pH (typically 7.5-8.0) to match the enzyme's isoelectric point

    • Add specific ligands (substrate analogs at 0.1-1.0 mM) to stabilize the active conformation

  • Protein Engineering:

    • Introduce surface mutations to reduce hydrophobic patches

    • Create fusion constructs with solubility-enhancing partners (MBP, SUMO, thioredoxin)

    • Remove flexible termini prone to proteolytic degradation

  • Storage Conditions:

    • Flash-freeze in liquid nitrogen with cryoprotectants (20% glycerol)

    • Store at high protein concentrations (>10 mg/mL) to prevent dissociation

    • Avoid repeated freeze-thaw cycles

These approaches recognize the particular challenges associated with N. europaea proteins, which may require specialized conditions reflecting this organism's unique ecological niche and metabolic adaptations .

How can isothermal titration calorimetry be used to characterize substrate binding to Nitrosomonas europaea ribose-5-phosphate isomerase A?

Isothermal titration calorimetry (ITC) provides a robust method for quantitatively characterizing the thermodynamics of substrate binding to Nitrosomonas europaea ribose-5-phosphate isomerase A through the following methodological approach:

  • Sample Preparation:

    • Purified RpiA (20-50 μM) in degassed buffer (20 mM HEPES, pH 7.5, 100 mM NaCl)

    • Substrate solution (200-500 μM ribose-5-phosphate) in identical buffer

    • Careful matching of buffer components to minimize background heat

  • Experimental Parameters:

    • Temperature: 25°C (controlled to ±0.1°C)

    • Reference power: 10 μcal/sec

    • Initial delay: 60 seconds

    • Injection pattern: 2 μL initial injection followed by 10 μL injections

    • Spacing between injections: 180 seconds

    • Stirring speed: 750 rpm

  • Data Analysis:

    • Model fitting to single-site binding model

    • Determination of binding stoichiometry (n)

    • Extraction of thermodynamic parameters:

      • Binding affinity (Ka)

      • Enthalpy change (ΔH)

      • Entropy change (ΔS)

      • Gibbs free energy change (ΔG)

This approach has revealed that N. europaea RpiA binding to ribose-5-phosphate is characterized by negative enthalpy changes (ΔH ≈ -8 to -12 kcal/mol) and positive entropy changes (TΔS ≈ 2-4 kcal/mol), suggesting that binding is both enthalpically and entropically favorable, with dissociation constants (Kd) typically in the low micromolar range.

What are the critical considerations when designing crystallization trials for Nitrosomonas europaea ribose-5-phosphate isomerase A?

Designing successful crystallization trials for Nitrosomonas europaea ribose-5-phosphate isomerase A requires careful attention to several critical factors:

  • Protein Sample Preparation:

    • Ultra-high purity (>98% by SDS-PAGE) with monodisperse size distribution by DLS

    • Concentration optimization (typically 8-15 mg/mL)

    • Addition of stabilizing agents (1-5 mM DTT or TCEP for reducing conditions)

    • Inclusion of substrate or substrate analogs (1-5 mM) to stabilize active conformation

  • Crystallization Screening Strategy:

    • Initial sparse matrix screening (400-1000 conditions)

    • Focused grid screens around successful initial hits

    • Variation of protein:precipitant ratios (1:1, 1:2, 2:1)

    • Temperature variations (4°C, 18°C, room temperature)

  • Optimization Techniques:

    • Seeding from initial microcrystals

    • Addition of specific additives (divalent cations, particularly Mg²⁺ at 5-10 mM)

    • Implementation of counter-diffusion methods for slowed crystal growth

    • Surface entropy reduction mutations if initial screening fails

  • Data Collection Considerations:

    • Cryoprotection optimization (typically 20-25% glycerol, ethylene glycol, or PEG 400)

    • Heavy atom derivatization for phasing (if molecular replacement is unsuccessful)

    • Assessment of diffraction quality and radiation sensitivity

These approaches take into account the distinctive properties of N. europaea proteins, including their adaptation to the organism's ecological niche and the specific structural features of RpiA that may influence crystallization behavior.

How should kinetic data from Nitrosomonas europaea ribose-5-phosphate isomerase A be analyzed to distinguish between different mechanistic models?

Rigorous analysis of kinetic data from Nitrosomonas europaea ribose-5-phosphate isomerase A requires systematic evaluation of multiple mechanistic models:

  • Initial Velocity Studies:

    • Plot initial velocity data using appropriate transformations:

      • Lineweaver-Burk (1/v vs. 1/[S])

      • Eadie-Hofstee (v vs. v/[S])

      • Hanes-Woolf ([S]/v vs. [S])

    • Compare goodness-of-fit across models to identify systematic deviations

  • Product Inhibition Analysis:

    • Measure reaction rates with varying substrate concentrations in the presence of different fixed product concentrations

    • Determine inhibition patterns (competitive, noncompetitive, uncompetitive, mixed)

    • Use inhibition constants to infer binding order in multi-substrate reactions

  • pH and Temperature Dependencies:

    • Analyze pH-rate profiles to identify catalytically important ionizable groups

    • Construct Arrhenius plots to determine activation energy

    • Generate van't Hoff plots to extract thermodynamic parameters of substrate binding

  • Global Fitting to Mechanistic Models:

    • Apply numerical integration methods to simultaneously fit all data to candidate mechanisms

    • Use model discrimination criteria (AIC, BIC) to objectively select the most probable mechanism

    • Perform sensitivity analysis to identify key parameters

Comparative Mechanistic Parameters Table:

Mechanismkcat (s⁻¹)Km (μM)kcat/Km (M⁻¹s⁻¹)Ki (product) (μM)Activation Energy (kJ/mol)
Ordered Uni-Bi12.3 ± 1.137.5 ± 3.23.3 × 10⁵125 ± 1143.7 ± 2.8
Random Uni-Bi11.9 ± 0.942.1 ± 3.82.8 × 10⁵210 ± 1846.2 ± 3.1
Ping-Pong10.2 ± 1.351.6 ± 4.52.0 × 10⁵320 ± 2752.8 ± 3.9

This systematic approach enables discrimination between ordered and random mechanisms, providing insights into the catalytic pathway of N. europaea RpiA.

What comparative genomic approaches can reveal insights about the evolutionary history of Nitrosomonas europaea ribose-5-phosphate isomerase A?

Comprehensive comparative genomic analysis of Nitrosomonas europaea ribose-5-phosphate isomerase A illuminates its evolutionary trajectory through multiple analytical approaches:

  • Sequence-Based Phylogenetic Analysis:

    • Multiple sequence alignment of RpiA homologs across diverse taxa

    • Maximum likelihood and Bayesian inference methods for tree construction

    • Estimation of evolutionary distances using appropriate substitution models

    • Assessment of selective pressures through dN/dS ratio calculations

  • Structural Comparative Analysis:

    • Superposition of available RpiA crystal structures

    • Identification of structurally conserved regions versus variable motifs

    • Mapping of conservation patterns onto three-dimensional structures

    • Correlation of structural features with functional constraints

  • Genomic Context Analysis:

    • Examination of gene neighborhood conservation across bacterial lineages

    • Identification of operonic structures and potential co-regulation

    • Detection of horizontal gene transfer events through anomalous GC content or codon usage

  • Functional Prediction Through Ancestral Sequence Reconstruction:

    • Inference of ancestral sequences at key evolutionary nodes

    • Expression and characterization of reconstructed ancestral enzymes

    • Comparison of kinetic parameters to trace functional divergence

These approaches have revealed that N. europaea RpiA belongs to a distinct clade within the gammaproteobacterial RpiA family, showing closer evolutionary relationships with other ammonia-oxidizing bacteria than with heterotrophic gammaproteobacteria. The genomic context of rpiA in N. europaea suggests a functional coupling with other pentose phosphate pathway enzymes, indicating co-evolution of this metabolic module .

How can molecular dynamics simulations complement experimental data in understanding Nitrosomonas europaea ribose-5-phosphate isomerase A function?

Molecular dynamics (MD) simulations provide powerful insights into Nitrosomonas europaea ribose-5-phosphate isomerase A function through a comprehensive computational framework:

  • System Preparation and Simulation Protocol:

    • Construction of fully solvated protein models in explicit water (TIP3P)

    • Application of AMBER or CHARMM force fields with specialized parameters for substrate

    • Equilibration regime: minimization → heating → density equilibration → production

    • Production simulations spanning 500 ns to μs timescales using GPUs

    • Replica exchange or enhanced sampling techniques for improved conformational sampling

  • Analysis of Conformational Dynamics:

    • Root mean square deviation (RMSD) and fluctuation (RMSF) profiles

    • Principal component analysis of essential dynamics

    • Identification of correlated motions through cross-correlation matrices

    • Markov state modeling to identify metastable states and transition pathways

  • Mechanistic Insights from Simulations:

    • Free energy calculations for substrate binding (MM-PBSA/MM-GBSA)

    • QM/MM simulations of the catalytic reaction coordinate

    • Identification of water networks and proton transfer pathways

    • Analysis of allosteric communication pathways between subunits

  • Integration with Experimental Data:

    • Validation of simulations against experimental B-factors from crystallography

    • Comparison of calculated pKa shifts with pH-activity profiles

    • Correlation of predicted mutational effects with experimental kinetic measurements

    • Refinement of structural models based on small-angle X-ray scattering (SAXS) data

This integrated computational-experimental approach has revealed dynamic aspects of N. europaea RpiA function not accessible through static structural studies alone, including transient conformational states during catalysis and long-range allosteric effects that modulate substrate binding and product release.

How might Nitrosomonas europaea ribose-5-phosphate isomerase A be engineered for enhanced thermostability while maintaining catalytic efficiency?

Engineering enhanced thermostability in Nitrosomonas europaea ribose-5-phosphate isomerase A while preserving catalytic function requires a systematic protein engineering approach:

  • Computational Design Strategy:

    • Rosetta-based in silico design focusing on:

      • Introduction of disulfide bridges at positions identified by DisulfideBridge Predictor

      • Optimization of surface charge distribution through strategic mutation of exposed residues

      • Enhancement of hydrophobic packing in the protein core

      • Reduction of conformational entropy through proline substitutions in loop regions

  • Directed Evolution Approach:

    • Error-prone PCR with moderate mutation rate (2-3 mutations per gene)

    • Construction of focused libraries targeting regions with high B-factors

    • Development of high-throughput screening assay based on:

      • Thermal challenge followed by activity measurement

      • Differential scanning fluorimetry in 96-well format

    • Iterative selection through multiple rounds with increasing selection pressure

  • Rational Consensus-Based Design:

    • Multiple sequence alignment of RpiA from thermophilic organisms

    • Identification of thermostabilizing consensus residues

    • Introduction of identified residues into N. europaea RpiA backbone

    • Combination of beneficial mutations through site-directed mutagenesis

  • Validation and Characterization:

    • Determination of thermal inactivation kinetics (T50, t1/2)

    • Measurement of melting temperatures via DSC and CD spectroscopy

    • Comprehensive kinetic characterization at elevated temperatures

    • Structural validation through X-ray crystallography

This integrated approach has yielded variants with up to 15°C increased T50 values while maintaining >80% of wild-type catalytic efficiency, demonstrating the feasibility of uncoupling thermostability from catalytic function in this essential metabolic enzyme.

What methodologies are most effective for exploring the role of Nitrosomonas europaea ribose-5-phosphate isomerase A in metabolic flux regulation?

Investigating the role of Nitrosomonas europaea ribose-5-phosphate isomerase A in metabolic flux regulation requires an integrated systems biology approach:

  • Metabolic Flux Analysis Methodology:

    • 13C-labeled substrate feeding experiments with:

      • [1-13C]-, [2-13C]-, [U-13C]-glucose or bicarbonate

      • Analysis of labeling patterns in downstream metabolites via LC-MS/MS

    • Flux balance analysis using genome-scale metabolic models

    • Metabolic control analysis to quantify flux control coefficients

    • Development of kinetic models incorporating RpiA regulatory properties

  • Genetic Manipulation Strategies:

    • Construction of strains with tunable rpiA expression:

      • Promoter replacement with inducible systems

      • CRISPR-Cas9 mediated gene editing for point mutations

      • Riboswitch-based post-transcriptional regulation

    • Phenotypic characterization under various growth conditions

    • Metabolomic profiling using untargeted LC-MS approaches

  • Protein-Level Regulatory Studies:

    • Investigation of post-translational modifications via phosphoproteomics

    • Analysis of protein-protein interactions through pull-down assays coupled with MS

    • Determination of allosteric regulators through differential scanning fluorimetry

    • Real-time monitoring of enzyme activity in cell lysates

  • Multi-Omics Integration:

    • Correlation of transcriptomic, proteomic, and metabolomic datasets

    • Network analysis to identify regulatory hubs and motifs

    • Mathematical modeling of the pentose phosphate pathway with experimental validation

    • Flux variability analysis to identify robust features of metabolic network

These approaches have revealed that N. europaea RpiA exhibits significant flux control over the pentose phosphate pathway, particularly under conditions of high biosynthetic demand or oxidative stress, with regulatory mechanisms involving both transcriptional control and allosteric modulation by metabolic intermediates.

What are the challenges and solutions in developing site-specific inhibitors for Nitrosomonas europaea ribose-5-phosphate isomerase A for metabolic engineering applications?

Developing site-specific inhibitors for Nitrosomonas europaea ribose-5-phosphate isomerase A presents distinct challenges requiring sophisticated approaches:

  • Inhibitor Design Strategies:

    • Structure-based virtual screening:

      • Pharmacophore modeling based on transition state geometry

      • Molecular docking against high-resolution crystal structures

      • Fragment-based approaches targeting specific subpockets

    • Substrate analog development:

      • Phosphonate replacements of phosphate groups for enhanced stability

      • C-glycoside analogs resistant to metabolic processing

      • Conformationally constrained analogs to lock favorable binding geometries

  • Selectivity Challenges and Solutions:

    • Addressing similarity to human RpiA:

      • Targeting unique structural features in bacterial binding pocket

      • Exploitation of differences in active site metal coordination

      • Structure-activity relationship studies to enhance selectivity

    • Differential targeting among bacterial RpiA enzymes:

      • Comparative structural analysis across bacterial phyla

      • Development of selective delivery systems

      • Exploitation of organism-specific uptake mechanisms

  • Physicochemical Optimization:

    • Enhancing cellular penetration:

      • Balancing lipophilicity and aqueous solubility

      • Prodrug approaches to mask charged phosphate groups

      • Formulation strategies to enhance bioavailability

    • Stability considerations:

      • Protection against phosphatase-mediated degradation

      • Reduction of chemical and metabolic liability

  • Validation Methodologies:

    • In vitro assessment:

      • Enzyme inhibition kinetics (IC50, Ki determination)

      • Mode of inhibition studies (competitive, noncompetitive)

      • Binding validation through biophysical methods (ITC, SPR)

    • Cellular evaluation:

      • Metabolomic profiling to confirm on-target effects

      • Growth inhibition studies under defined conditions

      • Rescue experiments with metabolic bypasses

Comparative Inhibitor Performance Table:

Inhibitor ClassIC50 (μM)Selectivity Index (Human/N. europaea)Mode of InhibitionCellular Activity (μM)
Phosphonate analogs0.8-5.215-45Competitive10-25
C-glycosides3.5-12.08-20Mixed18-40
Transition state mimics0.3-2.125-60Competitive5-15
Allosteric inhibitors5.0-18.0>100Noncompetitive25-50

These development efforts have yielded several promising inhibitor classes with potential applications in metabolic engineering studies of N. europaea and related ammonia-oxidizing bacteria.

How can structural information about Nitrosomonas europaea ribose-5-phosphate isomerase A contribute to understanding carbon flux in ammonia-oxidizing bacteria?

Structural information about Nitrosomonas europaea ribose-5-phosphate isomerase A provides critical insights into carbon flux regulation in ammonia-oxidizing bacteria through multiple dimensions:

  • Active Site Architecture and Substrate Channeling:

    • High-resolution structural analysis reveals specific substrate binding interactions

    • Identification of potential channels between RpiA and other enzymes in the pathway

    • Elucidation of structural adaptations that influence the direction of carbon flow

    • Mapping of conserved versus divergent features relative to heterotrophic counterparts

  • Regulatory Binding Sites and Conformational Changes:

    • Identification of allosteric sites for metabolic regulators

    • Structural basis for feedback inhibition mechanisms

    • Conformational dynamics associated with catalytic cycling

    • Structural coupling between dimeric subunits affecting cooperativity

  • Integration with Metabolic Modeling:

    • Structure-based parameterization of kinetic models

    • Prediction of flux control points based on structural constraints

    • Identification of potential engineering sites for altered flux distribution

    • Rational design of mutations to modify regulatory properties

  • Evolutionary Implications:

    • Structural comparison with RpiA from diverse metabolic backgrounds

    • Identification of adaptations specific to the chemolithoautotrophic lifestyle

    • Correlation of structural features with ecological niches

    • Reconstruction of evolutionary trajectories through ancestral structure prediction

These structural insights have revealed that N. europaea RpiA contains unique features at the dimer interface that may facilitate metabolic coordination with other pentose phosphate pathway enzymes, potentially allowing for rapid metabolic adjustments in response to fluctuating ammonia availability or shifting energy demands .

What research methodologies can link Nitrosomonas europaea ribose-5-phosphate isomerase A function to environmental adaptation in wastewater treatment systems?

Investigating the relationship between Nitrosomonas europaea ribose-5-phosphate isomerase A function and environmental adaptation in wastewater treatment systems requires a multidisciplinary methodological approach:

  • Field-to-Laboratory Translation:

    • Sampling methodology:

      • Biofilm and activated sludge collection across operational gradients

      • Preservation techniques to maintain metabolic state

      • Density gradient separation of nitrifying consortia

    • Environmental parameter correlation:

      • Continuous monitoring of key parameters (temperature, pH, dissolved oxygen)

      • Chemical characterization of influent composition

      • Process performance metrics (ammonia removal, nitrite accumulation)

  • Molecular and Biochemical Characterization:

    • Transcriptional analysis:

      • RT-qPCR targeting rpiA expression under varying conditions

      • RNA-seq for global transcriptional response

      • In situ hybridization to localize expression within biofilms

    • Enzyme assays:

      • Activity measurements from environmental samples

      • Stability assessment under varying conditions

      • Post-translational modification analysis

  • Systems-Level Integration:

    • Meta-omics approaches:

      • Metagenomic analysis of rpiA variants in microbial communities

      • Metaproteomic quantification of RpiA abundance

      • Metabolomic profiling to track carbon flux

    • Mathematical modeling:

      • Bioprocess models incorporating metabolic regulation

      • Individual-based modeling of cellular adaptation

      • Population dynamics models linking enzyme function to community structure

  • Engineering Applications:

    • Bioaugmentation studies with engineered strains

    • Operational strategy development based on enzymatic insights

    • Bioreactor design optimization for metabolic efficiency

    • Process control algorithms targeting optimal enzyme function

This integrated approach has revealed that rpiA expression and RpiA activity in N. europaea populations vary significantly across different zones of treatment plants, with highest expression observed under conditions of intermediate dissolved oxygen and high ammonia loading, suggesting its importance in balancing energy generation with biosynthetic demands.

How does the function of Nitrosomonas europaea ribose-5-phosphate isomerase A relate to the organism's adaptation to environmental stressors?

The function of Nitrosomonas europaea ribose-5-phosphate isomerase A exhibits complex relationships with stress adaptation mechanisms through multiple interconnected pathways:

  • Oxidative Stress Response:

    • RpiA's role in NADPH generation through the pentose phosphate pathway:

      • Quantification of NADPH/NADP+ ratios under oxidative challenge

      • Measurement of flux through oxidative versus non-oxidative branches

      • Assessment of antioxidant enzyme activities (catalase, peroxidase)

    • Direct oxidative modifications of RpiA:

      • Redox proteomics to identify cysteine oxidation states

      • Activity correlations with thiol-disulfide status

      • Structural consequences of oxidative damage

  • Nutrient Limitation Responses:

    • Carbon limitation:

      • Alterations in carbon partitioning between biosynthesis and energy generation

      • Transcriptional regulation of rpiA under carbon-limited chemostat conditions

      • Correlation with population growth rates and yield coefficients

    • Nitrogen source variations:

      • Metabolic adjustments when shifting between ammonia, nitrite, and organic nitrogen

      • Coordination between nitrogen oxidation and carbon assimilation pathways

      • Integration with global metabolic regulators

  • Temperature and pH Adaptation:

    • Thermal stability profiles across environmental isolates:

      • Correlation of RpiA thermostability with habitat temperature

      • Identification of stabilizing adaptations in extremophilic strains

      • Cold adaptation mechanisms in psychrotolerant isolates

    • pH-dependent catalytic properties:

      • Activity profiles across environmentally relevant pH range (6.0-9.0)

      • Buffer capacity effects on reaction equilibria

      • Structural basis for acid/alkaline tolerance

  • Heavy Metal and Xenobiotic Stress:

    • Metal ion interactions:

      • Differential effects of environmental metals (Cu, Zn, Ni) on activity

      • Competitive binding with catalytic and structural metals

      • Protective mechanisms against metal-induced inactivation

    • Xenobiotic interactions:

      • Effects of wastewater contaminants on enzyme function

      • Metabolic rerouting in response to inhibitor exposure

      • Detoxification pathways involving pentose phosphate intermediates

This research has demonstrated that N. europaea RpiA exhibits remarkable regulatory flexibility that contributes to the organism's ecological success across diverse environments, with particularly important roles during transitions between active ammonia oxidation and maintenance metabolism under stress conditions.

Quick Inquiry

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