Recombinant Rhodopirellula baltica Probable Nicotinate-Nucleotide Adenylyltransferase (nadD) is a recombinant protein derived from the marine bacterium Rhodopirellula baltica. This enzyme is involved in the biosynthesis of NAD+, playing a crucial role in cellular metabolism by converting nicotinate mononucleotide into NAD+ through adenylylation. The recombinant form of this enzyme is produced in E. coli and has been studied for its biochemical properties and potential applications.
Function: The nadD enzyme, also known as Nicotinate-Nucleotide Adenylyltransferase, catalyzes the adenylylation of nicotinate mononucleotide to form NAD+, which is essential for various cellular processes including energy metabolism and DNA repair.
Expression and Purification: The recombinant nadD enzyme is expressed in E. coli and purified to a high degree of purity (>85% as determined by SDS-PAGE) . The enzyme's sequence is well-defined, starting with MSASETTPQS and ending with YRVR, indicating a full-length protein of 214 amino acids .
Storage and Stability: The enzyme is stable for 6 months in liquid form and 12 months in lyophilized form when stored at -20°C or -80°C. Repeated freezing and thawing should be avoided, and working aliquots can be stored at 4°C for up to one week .
Metabolic Engineering: The nadD enzyme could be used in metabolic engineering strategies to enhance NAD+ biosynthesis in industrial microorganisms, improving their productivity and stress tolerance.
Pharmaceutical Applications: Given its role in NAD+ biosynthesis, nadD could be explored for applications in pharmaceuticals, particularly in areas related to energy metabolism and aging.
Recombinant Rhodopirellula baltica Probable Nicotinate-Nucleotide Adenylyltransferase (nadD) is a valuable enzyme for understanding NAD+ biosynthesis and its applications in biotechnology. Its high purity and stability make it suitable for various biochemical studies and potential industrial applications. Further research could explore its structural properties and biotechnological uses more deeply.
KEGG: rba:RB8455
STRING: 243090.RB8455
Nicotinate-nucleotide adenylyltransferase (nadD) is an essential enzyme in the biosynthesis of the NAD cofactor. In R. baltica, as in other organisms, nadD catalyzes the conversion of nicotinic acid mononucleotide (NaMN) to nicotinic acid adenine dinucleotide (NaAD) using ATP as a substrate. This reaction represents a critical step in the NAD biosynthetic pathway, which is essential for numerous cellular processes including energy metabolism, signal transduction, and redox reactions . In R. baltica, nadD likely plays a crucial role in the organism's adaptation to changing environmental conditions, particularly during transitions between growth phases, as suggested by the differential expression of metabolic and biosynthetic enzymes observed during life cycle analysis .
While specific structural data for R. baltica nadD is not yet fully characterized in the available literature, comparative analysis with the well-studied Mycobacterium tuberculosis nadD (MtNadD) provides valuable insights. Both enzymes belong to the nucleotidyltransferase superfamily of dinucleotide binding α/β-phosphodiesterases, characterized by a Rossmann fold with six-stranded parallel β sheets flanked by helices and connecting loops .
A notable distinction may exist in regulatory mechanisms. For instance, MtNadD exhibits an "over-closed" conformation imposed by a tightly packed α-helical segment, which differs from the extended coil commonly found in other NadD structures. This unique conformational feature may regulate enzymatic activity by transitioning between closed (inactive) and open (active) states in response to ATP levels . It would be valuable to investigate whether R. baltica nadD possesses similar regulatory mechanisms, particularly given R. baltica's environmental adaptability.
Life cycle analysis of R. baltica reveals complex transcriptional regulation patterns during different growth phases. While specific nadD expression patterns have not been directly reported, related metabolic enzymes show differential expression across growth phases. For example, during the transition from exponential to stationary phase, R. baltica increases expression of genes associated with energy production, amino acid biosynthesis, signal transduction, and stress response, while repressing genes involved in carbon metabolism and translation .
In particular, the stationary phase shows upregulation of genes for ubiquinone biosynthesis (RB2748, RB2749, and RB2750), which may be relevant to nadD function since both pathways relate to redox metabolism . This suggests that nadD expression might be coordinated with other metabolic changes during R. baltica's adaptation to nutrient limitation or environmental stress.
When designing experiments to study R. baltica nadD enzyme kinetics, researchers should follow these methodological steps:
Experimental objective definition: Clearly define whether you are studying basic kinetic parameters (Km, Vmax, kcat), inhibitor effects, substrate specificity, or regulatory mechanisms .
Selection of appropriate response variables: For nadD studies, consider measuring:
Factor selection and experimental design: Implement a factorial or response surface methodology (RSM) design to systematically evaluate factors such as:
The table below provides an example of a two-factor RSM design for studying pH and temperature effects on nadD activity:
| Run | pH | Temperature (°C) | nadD Activity (nmol/min/mg) |
|---|---|---|---|
| 1 | 6.5 | 25 | To be determined |
| 2 | 7.0 | 25 | To be determined |
| 3 | 7.5 | 25 | To be determined |
| 4 | 6.5 | 30 | To be determined |
| 5 | 7.0 | 30 | To be determined |
| 6 | 7.5 | 30 | To be determined |
| 7 | 6.5 | 35 | To be determined |
| 8 | 7.0 | 35 | To be determined |
| 9 | 7.5 | 35 | To be determined |
| 10 | 7.0 | 30 | To be determined |
For kinetic mechanism determination, consider implementing systematic varied-substrate concentration matrices to distinguish between sequential and ping-pong mechanisms, similar to approaches used for MtNadD characterization .
Changes in experimental design during R. baltica nadD studies are not uncommon due to technological advances, resource constraints, or unexpected findings. Researchers should follow these methodological steps to maintain scientific rigor:
A robust approach is to develop a predictive model from initial data, then test specific predictions in subsequent phases. This approach transforms what might appear as ad hoc design modifications into a more structured, hypothesis-driven investigation .
The choice of expression system for recombinant R. baltica nadD should be carefully considered based on experimental objectives. While specific data for R. baltica nadD expression is limited, insights can be drawn from successful approaches with homologous proteins like MtNadD:
E. coli expression systems:
BL21(DE3) strains with pET-based vectors have proven effective for expressing soluble NadD proteins
Consider N-terminal fusion tags (His6-tag with TEV protease cleavage site) to facilitate purification without compromising activity
Expression conditions: Typically, induction with IPTG (0.2-0.5 mM) at lower temperatures (16-25°C) for 16-20 hours provides better solubility
Purification strategy:
Quality assessment:
For structural studies, consider site-directed mutagenesis to enhance protein expression or stability. For example, with MtNadD, the W117A mutant showed approximately 10-fold higher expression levels compared to wild-type protein while maintaining structural integrity .
The regulatory mechanism of R. baltica nadD likely involves conformational changes that modulate enzymatic activity, similar to but potentially distinct from the mechanism observed in MtNadD. In MtNadD, a unique "over-closed" conformation rendered by a 3₁₀ helix locks the active site in a catalytically incompetent state that is topologically incompatible with substrate binding. This appears to represent a novel regulatory mechanism potentially triggered by low ATP levels .
To investigate whether R. baltica nadD employs a similar regulatory strategy, researchers should consider:
Structural characterization: Determine the crystal structure of R. baltica nadD in apo and substrate-bound forms to identify potential conformational states. Compare these with the known structures of MtNadD and other bacterial NadD proteins.
Site-directed mutagenesis: Target residues in potential regulatory elements (such as the equivalent region to the MtNadD 3₁₀ helix) to assess their impact on enzymatic activity and conformational dynamics.
Kinetic analysis under varying ATP conditions: Evaluate whether R. baltica nadD activity shows sensitivity to ATP concentration in a manner suggesting conformational regulation, particularly in environments mimicking the marine conditions where R. baltica naturally occurs.
Molecular dynamics simulations: Model the conformational transitions between potential active and inactive states to understand the energetics and triggers for such transitions.
This investigation would not only enhance our understanding of R. baltica nadD but could also reveal evolutionary adaptations in enzyme regulation across different bacterial species occupying distinct ecological niches .
R. baltica demonstrates remarkable adaptability to changing environmental conditions, particularly during transitions between growth phases. The relationship between nadD activity and these adaptive responses represents a complex and important research area:
Growth phase-dependent regulation: During the transition from exponential to stationary phase, R. baltica upregulates genes associated with energy production, amino acid biosynthesis, and stress response while downregulating genes involved in carbon metabolism and translation . The regulation of nadD likely integrates with these broader metabolic shifts, particularly those affecting NAD(P) homeostasis.
Cell wall modifications: R. baltica modifies its cell wall composition in response to environmental changes, including increased production of polysaccharides and formation of rosettes in stationary phase, suggesting production of holdfast substances . The NAD cofactor produced through the nadD pathway may support these structural adaptations through its role in redox reactions and signaling.
Integration with oxygen sensing: R. baltica increases production of ubiquinone in response to oxygen limitation during stationary phase . Since both NAD and ubiquinone pathways are involved in electron transport and redox balance, nadD activity may be coordinated with oxygen-sensing mechanisms.
Methodology for investigation: To elucidate these relationships, researchers should consider:
Transcriptomic and proteomic profiling across growth phases and environmental conditions
Metabolic flux analysis focused on NAD(P) pathways
Genetic modification approaches (if available for R. baltica) to modulate nadD expression
Correlation of nadD activity with specific adaptive phenotypes
Understanding these relationships could provide insights into how R. baltica has evolved to thrive in its marine environment and how NAD metabolism contributes to bacterial adaptation more broadly.
The evolutionary conservation of nadD across Planctomycetes represents a valuable molecular marker for understanding phylogenetic relationships within this unique bacterial phylum. This investigation requires multi-faceted approaches:
Sequence comparison and phylogenetic analysis:
Conduct comprehensive sequence alignments of nadD genes from diverse Planctomycetes
Construct phylogenetic trees using maximum likelihood or Bayesian methods
Compare nadD-based phylogenies with those derived from 16S rRNA and other conserved genes
Identify signature motifs or domains specific to Planctomycetes nadD proteins
Structural conservation analysis:
Evaluate conservation of key structural elements across Planctomycetes nadD proteins
Assess whether unique structural features (like regulatory elements) are conserved within specific clades
Correlate structural conservation with ecological niches or metabolic capabilities
Functional divergence investigation:
Compare substrate specificities and kinetic parameters across different Planctomycetes
Identify adaptations in catalytic or regulatory mechanisms that correlate with specific environmental conditions
Assess whether key differences exist between marine Planctomycetes like R. baltica and those from other habitats
Horizontal gene transfer assessment:
Evaluate evidence for horizontal acquisition of nadD genes within Planctomycetes
Identify potential genetic exchange with other bacterial phyla
Planctomycetes are notable for their unusual cell biology, including membrane-enclosed nucleoids and proteinaceous cell walls instead of peptidoglycan . Understanding the evolution of key metabolic enzymes like nadD can provide insights into how these unique features evolved and how they relate to the group's distinct ecological roles in marine and other environments.
When analyzing kinetic data from R. baltica nadD enzyme assays, researchers should implement a multi-tiered statistical approach:
Nonlinear regression for kinetic parameter estimation:
For initial velocity studies, fit data to appropriate enzyme kinetic equations using nonlinear regression rather than linear transformations (e.g., Lineweaver-Burk plots), which can distort error structure
For a sequential bi-substrate mechanism (likely for nadD), use the appropriate rate equation:
Include appropriate weighting schemes based on the error structure of your measurements
Model discrimination:
Experimental design optimization:
Robust statistical analysis:
The table below illustrates a systematic approach to varying substrates for mechanism determination:
| [NaMN] (μM) | [ATP] (μM) | Initial Velocity (μmol/min/mg) |
|---|---|---|
| 50 | 50 | v₁ |
| 50 | 100 | v₂ |
| 50 | 200 | v₃ |
| 100 | 50 | v₄ |
| 100 | 100 | v₅ |
| 100 | 200 | v₆ |
| 200 | 50 | v₇ |
| 200 | 100 | v₈ |
| 200 | 200 | v₉ |
Analysis of these data using appropriate nonlinear regression would allow discrimination between ordered, random, and ping-pong mechanisms, similar to approaches used for MtNadD characterization .
Contradictory results are not uncommon in enzyme characterization studies and may arise from methodological differences, environmental conditions, or inherent properties of the enzyme. Researchers should employ a systematic approach to resolve such discrepancies:
Source identification and documentation:
Controlled comparative studies:
Multiple orthogonal methods:
Employ different analytical techniques to measure the same property
For example, study protein-substrate interactions using both kinetic assays and biophysical methods like isothermal titration calorimetry (ITC) or surface plasmon resonance (SPR)
Compare recombinant nadD from different expression systems to rule out tag or purification artifacts
Statistical meta-analysis:
Biological explanation exploration:
Understanding contradictions often leads to deeper insights into enzyme mechanisms. For example, the discovery of the closed conformation in MtNadD initially appeared contradictory to its enzymatic activity but ultimately revealed an important regulatory mechanism .
Developing comprehensive models of R. baltica nadD regulation requires effective integration of multi-omics data through a systems biology approach:
Data collection and standardization:
Generate or compile structural data (X-ray crystallography, cryo-EM) of R. baltica nadD in different conformational states
Perform functional assays under varying conditions that mimic R. baltica's natural environment
Analyze transcriptomic and proteomic data across growth phases and environmental conditions
Multi-level integration methodology:
Use protein structure networks to map how conformational changes affect enzyme activity
Correlate transcriptomic changes with metabolic flux through the NAD biosynthetic pathway
Apply machine learning approaches to identify patterns in large datasets that may not be apparent through traditional analysis
Implement Bayesian network models to infer causal relationships between environmental factors and nadD regulation
Regulatory network construction:
Validation through targeted experiments:
Dynamic modeling:
By integrating these diverse data types, researchers can develop models that explain how R. baltica regulates nadD activity in response to changing environmental conditions, providing insights into both the specific enzyme mechanism and broader bacterial adaptation strategies.
Designing effective inhibitor screens for R. baltica nadD requires careful consideration of multiple factors to ensure scientific rigor and relevance:
Assay development and validation:
Establish a robust, reproducible activity assay suitable for high-throughput screening
Consider coupled enzymatic assays that monitor either ATP consumption or NaAD formation
Validate assay parameters: Z' factor >0.5, signal-to-background ratio >5, and coefficient of variation <10%
Include appropriate positive controls (known inhibitors of related enzymes) and negative controls
Compound library design considerations:
Focus on chemical scaffolds known to interact with nucleotidyltransferases
Include compounds targeting the unique structural features of nadD
Consider fragment-based approaches to identify novel binding modes
Incorporate natural products, particularly from marine environments relevant to R. baltica's ecology
Screening strategy optimization:
Structure-activity relationship analysis:
Correlate inhibitory potency with chemical features using quantitative structure-activity relationship (QSAR) models
Use structural information from homologous enzymes (like MtNadD) to guide rational optimization of hits
Consider molecular docking and molecular dynamics simulations to predict binding modes
Selectivity assessment:
The table below provides a framework for categorizing inhibitors based on mechanism and selectivity:
| Inhibitor Type | Mechanism | Advantages | Challenges | Examples |
|---|---|---|---|---|
| Competitive (NaMN site) | Blocks substrate binding | Often high potency | May lack selectivity | Nucleotide analogs |
| Competitive (ATP site) | Blocks ATP binding | Well-established approaches | Cross-reactivity with other ATP-utilizing enzymes | ATP-mimetics |
| Allosteric | Stabilizes inactive conformation | Potentially higher selectivity | May be difficult to identify | Small molecules targeting regulatory sites |
| Covalent | Forms chemical bond with enzyme | Long-lasting inhibition | Potential off-target effects | Electrophilic compounds |
This systematic approach will help identify inhibitors with potential applications in basic research and possibly as scaffolds for antibacterial development against related pathogens .
Determining the oligomeric state of recombinant R. baltica nadD is crucial for understanding its structure-function relationships. Researchers should employ multiple complementary approaches:
Size exclusion chromatography (SEC):
Run purified nadD on analytical SEC columns calibrated with molecular weight standards
Compare elution profiles under different conditions (salt concentration, pH, presence of substrates)
Analyze SEC data using calibration curves to estimate apparent molecular weight
Consider the limitation that shape (Stokes radius) affects elution behavior
Multi-angle light scattering (MALS):
Analytical ultracentrifugation (AUC):
Native mass spectrometry:
Use native electrospray ionization mass spectrometry to determine intact complex mass
Evaluate stability of oligomers under different solution conditions
Identify potential cofactors or ligands co-purifying with the complex
Structural analysis:
Functional correlation:
Based on homologous NadD proteins, R. baltica nadD likely functions as a dimer, though tetramer formation might occur under specific conditions such as high protein concentration or presence of interface-stabilizing ligands . Understanding the relationship between oligomerization and function could reveal important regulatory mechanisms specific to R. baltica's adaptation to its marine environment.
Investigation of potential post-translational modifications (PTMs) of R. baltica nadD requires a comprehensive strategy utilizing cutting-edge proteomics techniques and functional correlation studies:
Mass spectrometry-based PTM identification:
Perform bottom-up proteomics analysis using high-resolution mass spectrometry
Implement multiple proteolytic enzymes (trypsin, chymotrypsin, Glu-C) to maximize sequence coverage
Use enrichment strategies for specific PTMs:
Phosphorylation: TiO₂ or IMAC enrichment
Acetylation: Anti-acetyllysine antibodies
Methylation: Anti-methyllysine/arginine antibodies
Apply electron transfer dissociation (ETD) or electron capture dissociation (ECD) for labile PTM characterization
Site-directed mutagenesis validation:
Functional correlation studies:
Identification of regulatory enzymes:
Integrative analysis:
Correlate PTM sites with structural features using available structural data
Map modifications onto key functional regions (substrate binding sites, oligomerization interfaces)
Compare with regulatory PTMs identified in related enzymes like human NAD kinase, where N- and C-terminal modifications have opposing regulatory effects
Recent research has revealed that terminal regions can significantly impact enzyme activity, as seen with human NAD kinase where the C-terminal region is critical for activity while the N-terminal region exhibits an inhibitory role . Investigation of similar regulatory mechanisms in R. baltica nadD could reveal unique adaptations to its marine environment.
Research on R. baltica nadD presents numerous promising avenues for investigation that could significantly advance our understanding of this enzyme, its regulatory mechanisms, and broader implications for bacterial metabolism:
Structural biology approaches:
Determine high-resolution structures of R. baltica nadD in multiple conformational states
Characterize substrate binding sites and potential allosteric regions
Apply cryo-EM to capture conformational dynamics not accessible by crystallography
Develop computational models of conformational transitions during the catalytic cycle
Systems biology integration:
Map the role of nadD within the broader NAD(P) metabolic network in R. baltica
Investigate how nadD activity coordinates with other aspects of cellular metabolism during environmental adaptation
Develop predictive models for how nadD regulation contributes to R. baltica's ecological fitness
Compare with NAD metabolism in other marine bacteria to identify unique adaptations
Evolutionary and comparative studies:
Conduct comprehensive phylogenetic analysis of nadD across diverse bacterial phyla
Investigate how nadD structure and function have evolved within Planctomycetes
Identify unique features that may reflect adaptation to marine environments
Explore potential horizontal gene transfer events involving nadD
Development of genetic tools:
Biotechnological applications:
These research directions would not only advance our fundamental understanding of NAD metabolism in Planctomycetes but could also provide insights into bacterial adaptation and potentially lead to biotechnological applications leveraging the unique properties of this marine enzyme.
Advancing R. baltica nadD research requires effective interdisciplinary collaboration that integrates diverse expertise and methodologies:
Building effective collaborative frameworks:
Integration of complementary methodologies:
Data sharing and management strategies:
Overcoming disciplinary barriers:
Organize workshops to build shared understanding of techniques and limitations
Rotate researchers between laboratories to transfer skills and perspectives
Develop "translator" roles for individuals with cross-disciplinary expertise
Establish common goals that address questions of interest to all participating disciplines
Leveraging institutional resources: