Recombinant Rhodopirellula baltica D-tyrosyl-tRNA (Tyr) deacylase (dtd) is an enzyme produced in yeast and encoded by the dtd gene in the marine bacterium Rhodopirellula baltica . This enzyme belongs to the D-aminoacyl-tRNA deacylase (DTD) family and is involved in removing D-amino acids that are mistakenly attached to transfer RNAs (tRNAs) . DTDs are thus crucial in maintaining the homochirality of proteins .
The primary function of D-tyrosyl-tRNA (Tyr) deacylase is to hydrolyze the ester bond between D-tyrosine and tRNA . This process is essential for removing incorrectly attached D-tyrosine from tRNA^{\text{Tyr}}\, ensuring that only L-amino acids are incorporated during protein synthesis. The enzyme's activity extends beyond chiral proofreading, as it also eliminates glycine mischarged to tRNA^{\text{Ala}}\ .
Involved in quality control during protein biosynthesis, DTD prevents the incorporation of D-amino acids into proteins, which could lead to non-functional or toxic products . By hydrolyzing D-tyrosyl-tRNA^{\text{Tyr}}\, the enzyme ensures that the correct L-tyrosine is used, maintaining the integrity of the genetic code during translation .
Rhodopirellula baltica, a marine bacterium, possesses this DTD, highlighting its importance in maintaining protein fidelity in diverse organisms . The genome analysis of R. baltica has revealed several biotechnologically relevant features, including unique sulfatases and C1-metabolism genes . Other organisms, such as Pirellula sp. strain 1, also possess DTD, minimizing the harmful effects of D-tyrosine binding to tyrosyl-tRNA .
The presence of unique enzymes and metabolic pathways in Rhodopirellula baltica suggests its potential in biotechnological applications . Further exploration of its enzymatic capabilities may reveal novel functions and applications in biocatalysis, bioremediation, and drug discovery.
KEGG: rba:RB8300
STRING: 243090.RB8300
D-tyrosyl-tRNA (Tyr) deacylase (DTD) in R. baltica likely functions as a quality control enzyme that prevents the incorporation of D-amino acids into proteins during translation. Similar to the human DTD1 enzyme, it hydrolyzes D-tyrosyl-tRNA(Tyr) into D-tyrosine and free tRNA(Tyr), serving as a defense mechanism against the potentially harmful effects of D-amino acids in protein synthesis . This function is particularly important for organisms like R. baltica that may encounter diverse environmental conditions in marine habitats, where D-amino acids could be present in sediments or produced by other microorganisms.
The enzyme belongs to a highly conserved family of proteins that play crucial roles in maintaining translational fidelity. While not explicitly characterized in the provided proteome studies of R. baltica, inferences can be made based on the conservation of this enzyme across different domains of life. The enzyme's importance in maintaining proper protein folding and function makes it an essential component of cellular protein quality control mechanisms.
Based on the genomic analysis of R. baltica, the DTD gene would be one of the 6,644 coding sequences identified in the genome annotation . The R. baltica genome has a size of approximately 7.4 Mb with a G+C content of 55.3% . While specific information about the DTD gene organization is not explicitly provided in the search results, it is likely located within the main chromosome, given that most housekeeping genes in bacteria tend to be chromosomally encoded.
The gene may be part of an operon related to translation or protein quality control functions, though this would require specific experimental verification through transcriptomic analysis. The genome annotation performed with PATRIC version 3.3.15 would provide the specific location and context of the DTD gene within the genome .
For expressing recombinant R. baltica DTD, researchers should consider several expression systems based on the properties of the protein and experimental needs:
E. coli expression system: Most commonly used for recombinant protein production due to its simplicity, rapid growth, and high yields. This system would be particularly suitable for R. baltica DTD given that bacterial proteins often express well in E. coli. Typical vectors would include pET series with IPTG-inducible promoters.
Native expression in R. baltica: For studying the protein in its natural context, expression in R. baltica itself could be considered. R. baltica can be grown in mineral medium with various carbon sources such as ribose, glucose, or N-acetylglucosamine . The cells would typically be harvested in exponential growth phase by centrifugation (10,000×g, 15 min, 4°C) .
Fusion tag selection: A C-terminal 6×His tag (similar to what's used for human DTD1 ) would facilitate purification through affinity chromatography. This approach would enable single-step purification while minimizing interference with enzyme activity.
The expression conditions should be optimized considering R. baltica's natural growth environment, which includes marine conditions. Protein extraction could follow protocols similar to those used in proteome studies of R. baltica, involving cell lysis in buffer containing urea, thiourea, DTT, CHAPS, and carrier ampholytes .
The structure-function relationship of R. baltica DTD likely shares conserved features with DTD enzymes from other species while possessing unique adaptations reflective of its marine environment:
Conserved features:
The catalytic mechanism likely involves key residues for recognizing the D-configuration of amino acids
The binding pocket probably maintains specificity for tyrosyl-tRNA
Structural elements for ATP binding may be present, as seen in human DTD1 which exhibits ATPase activity
Potential unique features:
Adaptations to marine conditions, possibly including salt tolerance mechanisms
Structural modifications that might enhance stability under the specific cellular compartmentalization in R. baltica
Possible adaptations related to the unusual cell morphology of R. baltica, which includes different compartments (pirellulosome containing the riboplasm, and paryphoplasm)
Comparative structural analysis would require protein crystallography studies of the recombinant R. baltica DTD. This could be achieved by purifying the protein with affinity chromatography using a His-tag system, followed by crystallization trials and X-ray diffraction analysis. The resulting structure could be compared with known DTD structures from other organisms to identify conserved motifs and species-specific adaptations.
A comprehensive purification protocol for recombinant R. baltica DTD would involve:
Expression optimization:
Clone the R. baltica DTD gene into an expression vector with a C-terminal 6×His tag
Transform into E. coli BL21(DE3) or similar expression strain
Optimize induction conditions (IPTG concentration, temperature, induction time)
Culture cells in LB or 2×YT medium at 25-30°C to enhance protein solubility
Cell lysis:
Affinity purification:
Apply clarified lysate to Ni-NTA column equilibrated with lysis buffer
Wash extensively with wash buffer (lysis buffer with 20 mM imidazole)
Elute with elution buffer (lysis buffer with 250 mM imidazole)
Secondary purification:
Apply eluted protein to size exclusion chromatography column (Superdex 75 or 200)
Collect fractions and analyze by SDS-PAGE
Pool pure fractions and concentrate using ultrafiltration
Activity preservation:
Add glycerol to final concentration of 10-20%
Store at -80°C in small aliquots to avoid freeze-thaw cycles
Include DTT or β-mercaptoethanol in storage buffer to maintain reduced state of cysteine residues
Enzyme activity should be monitored throughout purification using a deacylation assay that measures the release of D-tyrosine from D-tyrosyl-tRNA(Tyr) substrates, using either radioactive substrates or HPLC-based methods for D-tyrosine detection.
Growth phase significantly impacts protein expression profiles in R. baltica, which likely extends to DTD expression and activity:
The number of regulated proteins (fold changes > |2|) increases from early stationary phase (10 proteins) to late stationary phase (179 proteins)
Growth phase transitions show opposing regulation of key metabolic pathways, including tricarboxylic acid cycle and oxidative pentose phosphate cycle
As a quality control enzyme, DTD might show differential expression patterns depending on growth phase
If DTD functions similarly to other protein quality control mechanisms, it might be upregulated during stationary phase when stress responses are activated
DTD activity might correlate with the alternative sigma factor sigmaH, which is upregulated in stationary phase and often associated with stress responses
Grow R. baltica cultures in mineral medium with ribose as carbon source
Harvest cells at different growth phases (early exponential, late exponential, early stationary, late stationary)
Analyze DTD protein levels using western blot with anti-DTD antibodies
Measure DTD enzyme activity in cell extracts from different growth phases
Compare with 2D-DIGE profiles to correlate DTD expression with other regulated proteins
R. baltica possesses a unique cellular compartmentalization that divides the cell into distinct regions:
The intracytoplasmic membrane encloses the pirellulosome, which contains the riboplasm with ribosome-like particles and the condensed nucleoid
The region between the intracytoplasmic and cytoplasmic membranes contains the paryphoplasm, which harbors some RNA but no ribosome-like particles
Protein biosynthesis likely only takes place in the riboplasm compartment
Given its function in translation quality control, DTD is most likely localized to the riboplasm compartment where protein synthesis occurs
The enzyme would need to be in proximity to ribosomes to efficiently hydrolyze misaminoacylated tRNAs
Similar to human DTD1, which is localized to both nucleus and cytoplasm , R. baltica DTD might also show dual localization patterns
Generate antibodies against recombinant R. baltica DTD or create a fluorescently tagged version
Perform immunogold electron microscopy to visualize DTD localization within the compartmentalized cell structure
Alternatively, perform subcellular fractionation to separate the paryphoplasm and pirellulosome compartments
Analyze fractions by western blotting to detect DTD
Correlate findings with the presence/absence of signal peptides, as 146 of the identified proteins in R. baltica contained predicted signal peptides suggesting translocation
Understanding DTD localization would provide insights into the spatial organization of translation quality control mechanisms in this uniquely compartmentalized bacterium.
Studying the kinetics of R. baltica DTD requires specialized techniques to measure its deacylation activity:
Synthesize D-tyrosyl-tRNA(Tyr) substrate by:
In vitro transcription of tRNA(Tyr) gene
Aminoacylation using D-tyrosyl-tRNA synthetase or chemical aminoacylation methods
Purification of aminoacylated tRNA by chromatography
Radiometric assay:
Use 14C or 3H-labeled D-tyrosine to prepare labeled D-tyrosyl-tRNA(Tyr)
Measure release of labeled D-tyrosine after incubation with DTD
Determine initial velocities at different substrate concentrations
Calculate Km and kcat values using Michaelis-Menten kinetics
HPLC-based assay:
Incubate DTD with D-tyrosyl-tRNA(Tyr)
Analyze reaction mixture by reverse-phase HPLC
Quantify released D-tyrosine using UV detection or fluorescence after derivatization
Plot reaction rates versus substrate concentrations
Coupled enzyme assay:
Design a coupled assay where D-tyrosine release is linked to a detectable signal
For example, couple with D-amino acid oxidase and horseradish peroxidase
Measure H2O2 production spectrophotometrically using a chromogenic substrate
Km for D-tyrosyl-tRNA(Tyr) substrate
kcat for deacylation reaction
Substrate specificity by testing other D-aminoacyl-tRNAs
Effects of pH, temperature, and ionic strength on enzyme activity, particularly considering R. baltica's marine environment
These approaches would provide comprehensive insights into the catalytic efficiency and substrate preferences of R. baltica DTD.
Differential proteomics offers powerful approaches to investigate DTD's role in R. baltica stress responses:
Culture R. baltica under various stress conditions:
D-amino acid stress (addition of D-tyrosine or D-amino acid mixture)
Oxidative stress (H2O2 exposure)
Temperature stress (heat shock or cold shock)
Nutrient limitation
pH stress (relevant to marine environments)
Apply two-dimensional difference gel electrophoresis (2D-DIGE) methodology:
Label control and stressed samples with different fluorescent dyes (Cy2, Cy3, Cy5)
Combine samples and separate by 2D electrophoresis
Analyze differential protein expression using specialized software
This methodology has been successfully applied to study growth phase-dependent protein changes in R. baltica
Identify regulated proteins by mass spectrometry:
Create protein interaction networks centered on DTD
Correlate DTD expression with other stress response proteins
Compare expression changes across different stress conditions
Identify potential functional partners of DTD under stress
This approach would reveal whether DTD is part of specific stress response pathways and how its expression correlates with other proteins involved in translation quality control or stress response mechanisms.
A multi-omics approach can be employed to comprehensively identify DTD-interacting partners:
Comparative genomics: Analyze DTD gene neighborhood across related species to identify conserved gene clusters that might function together
Transcriptomic analysis: Perform RNA-Seq to identify genes co-expressed with DTD under various conditions
Genome-wide CRISPR interference: Systematically knock down genes and screen for effects on DTD function or expression
Affinity purification-mass spectrometry (AP-MS):
Express His-tagged or FLAG-tagged DTD in R. baltica
Perform gentle cell lysis to preserve protein-protein interactions
Capture DTD and associated proteins using affinity chromatography
Identify interacting proteins by LC-MS/MS
Distinguish true interactors from background using controls
Proximity-dependent biotin identification (BioID):
Fuse DTD to a biotin ligase (BirA*)
Express in R. baltica
Biotin-label proximal proteins in vivo
Purify biotinylated proteins using streptavidin beads
Identify by mass spectrometry
Cross-linking mass spectrometry (XL-MS):
Treat R. baltica cells with chemical cross-linkers
Enrich for DTD-containing complexes
Digest and analyze by specialized mass spectrometry
Identify cross-linked peptides that reveal direct interactions
Create interaction networks combining evidence from all approaches
Assign confidence scores based on detection across multiple methods
Validate key interactions using targeted approaches like co-immunoprecipitation or yeast two-hybrid
This integrated approach would provide a comprehensive view of the DTD interactome in R. baltica, revealing both stable and transient interactions that contribute to its function in translation quality control.
R. baltica DTD likely exhibits both conserved features and unique characteristics compared to DTD enzymes from other organisms:
| Aspect | R. baltica DTD (predicted) | E. coli DTD | Other bacterial DTDs |
|---|---|---|---|
| Domain structure | Single domain with conserved DTD fold | Single domain | Single domain |
| Substrate specificity | Likely D-tyrosyl-tRNA(Tyr) with possible broader specificity | Broad specificity for D-aminoacyl-tRNAs | Variable specificity |
| Catalytic mechanism | Hydrolysis without cofactors | Hydrolysis without cofactors | Generally cofactor-independent |
| Size | ~15-18 kDa (predicted) | ~15 kDa | 14-18 kDa |
| Environmental adaptation | Potential adaptations to marine conditions | Mesophilic adaptation | Variable based on habitat |
Human DTD1 is both nuclear and cytoplasmic , while R. baltica DTD is likely confined to the riboplasm compartment
Human DTD1 shows ATPase activity and involvement in DNA replication , functions not typically associated with bacterial DTDs
Eukaryotic DTDs often have additional domains or regulatory regions absent in bacterial counterparts
Human DTD1 is preferentially phosphorylated in cells arrested in S phase , representing a regulatory mechanism likely absent in R. baltica
DTD is a highly conserved enzyme across all domains of life, suggesting its fundamental importance
R. baltica belongs to the phylum Planctomycetes, which has a unique evolutionary position
Comparative genomic analysis could reveal whether R. baltica DTD shows unique adaptations related to its unusual cellular compartmentalization
These differences would have implications for experimental design when studying R. baltica DTD, particularly regarding assay conditions, substrate selection, and interpretation of functional data.
R. baltica's marine environment presents unique challenges that may influence DTD function and importance:
Marine environments can contain D-amino acids from bacterial cell walls and algal peptides
Fluctuating salinity may impact protein folding and stability, potentially increasing translational errors
Temperature variations in marine habitats could affect tRNA charging fidelity
Enhanced quality control: DTD may be particularly important for preventing incorporation of environmental D-amino acids into proteins
Stress response: DTD could play a role in R. baltica's ability to survive in changing marine conditions
Nutrient recycling: By reclaiming misaminoacylated tRNAs, DTD may contribute to efficient resource utilization in potentially nutrient-limited environments
R. baltica possesses genes associated with aromatic compound metabolism , suggesting adaptations to utilizing diverse carbon sources
The complex cell morphology of R. baltica, including motile swarmer cells and sessile rosettes , may require specialized quality control mechanisms during differentiation
Growth phase-dependent regulation of 179 proteins indicates complex adaptations to environmental changes
Compare DTD expression and activity under different salinity, temperature, and nutrient conditions
Assess DTD importance for survival when R. baltica is exposed to environmental D-amino acids
Examine whether DTD is differentially expressed in different R. baltica morphotypes (swarmer cells vs. rosettes)
Understanding DTD's role in environmental adaptation could provide insights into the molecular mechanisms underlying R. baltica's ecological success in marine habitats.
Comprehensive analysis of R. baltica DTD substrate specificity requires multiple complementary approaches:
Generate a diverse set of D-aminoacyl-tRNAs including:
D-tyrosyl-tRNA(Tyr) (primary predicted substrate)
Other D-aromatic-aminoacyl-tRNAs (D-Phe, D-Trp)
D-aliphatic-aminoacyl-tRNAs (D-Ala, D-Val, D-Leu)
D-charged-aminoacyl-tRNAs (D-Lys, D-Glu)
Prepare substrates using either:
Chemical aminoacylation with activated D-amino acids
Enzymatic aminoacylation using engineered aminoacyl-tRNA synthetases
Parallel reaction monitoring:
Incubate DTD with a mixture of different D-aminoacyl-tRNAs
Analyze reaction products by LC-MS/MS
Quantify deacylation rates for each substrate simultaneously
Calculate relative specificity constants (kcat/Km)
Thin-layer chromatography (TLC) assay:
Use radioactively labeled D-amino acids
Separate reaction products by TLC
Quantify using phosphorimager
Compare relative deacylation efficiencies
Real-time fluorescence assay:
Label tRNAs with fluorescent dyes
Monitor conformational changes during deacylation in real-time
Calculate reaction rates for different substrates
Create a substrate specificity profile showing relative activity against different D-aminoacyl-tRNAs
Determine kinetic parameters (Km, kcat, kcat/Km) for each substrate
Correlate specificity with amino acid properties (size, hydrophobicity, charge)
This comprehensive approach would provide detailed insights into the substrate preferences of R. baltica DTD, which could be related to its ecological niche and the types of D-amino acids it might encounter in its marine environment.
High-throughput screening (HTS) for DTD modulators can be implemented using the following systematic approach:
Fluorescence-based primary assay:
Develop a fluorogenic substrate (e.g., D-tyrosyl-tRNA(Tyr) with fluorescence quenched by aminoacylation)
Deacylation by DTD results in measurable fluorescence increase
Optimize in 384 or 1536-well format for miniaturization
Validate with known controls (heat-inactivated enzyme, varying enzyme concentrations)
Assay quality parameters:
Achieve Z' factor >0.5 for statistical reliability
Signal-to-background ratio >10
Coefficient of variation <10%
DMSO tolerance up to 1% for compound solubility
Primary screen:
Test compounds at single concentration (10-20 μM)
Include positive controls (known inhibitors if available) and negative controls
Set thresholds for hit selection (>50% inhibition or >150% enhancement)
Secondary validation:
Counterscreen against related hydrolases to assess selectivity
Dose-response curves to determine IC50 or EC50 values
Rule out interference mechanisms (fluorescence quenching, aggregation)
Tertiary characterization:
Determine mechanism of inhibition (competitive, non-competitive)
Assess reversibility
Evaluate effects in cellular context (if cellular system available)
| Parameter | Weight | Scoring criteria |
|---|---|---|
| Potency | 30% | IC50/EC50 value ranges |
| Selectivity | 25% | Activity against related enzymes |
| Mechanism | 20% | Competitive vs. allosteric |
| Chemical properties | 15% | Solubility, stability, synthetic accessibility |
| Novelty | 10% | Structural uniqueness |
This screening approach would identify chemical tools to study DTD function and potentially lead to compounds for investigating DTD's role in R. baltica physiology and adaptation.
Future research on R. baltica DTD should focus on several key areas that would advance our understanding of its biological significance:
Functional genomics approach:
Generate DTD knockout or depletion strains in R. baltica
Perform phenotypic characterization under various growth conditions
Analyze effects on proteome quality and D-amino acid tolerance
This would establish the importance of DTD for R. baltica survival and adaptation
Structural biology investigations:
Determine crystal structure of R. baltica DTD
Compare with structures from other organisms
Identify unique structural features that might relate to marine adaptation
Perform structure-guided mutagenesis to identify key catalytic residues
Systems biology integration:
Map the position of DTD in R. baltica's protein quality control network
Integrate proteomics, transcriptomics, and metabolomics data
Develop computational models of translation quality control
This would place DTD in the broader context of cellular homeostasis
Evolutionary studies:
Compare DTD sequences and functions across Planctomycetes
Investigate horizontal gene transfer events that might have shaped DTD evolution
Reconstruct the evolutionary history of DTD in relation to cellular compartmentalization
Environmental adaptation mechanisms:
These research directions would provide comprehensive insights into DTD's biological significance in R. baltica and potentially reveal novel aspects of translation quality control in bacteria with complex cellular organizations.
Recombinant R. baltica DTD offers several promising biotechnology applications based on its predicted enzymatic properties:
Biocatalysis and chiral chemistry:
Development of enzymatic methods for D-amino acid purification
Creation of stereospecific biocatalysts for pharmaceutical synthesis
DTD could be used to selectively remove D-amino acids from mixtures
Protein production quality control:
Addition of recombinant DTD to cell-free protein synthesis systems
Reduction of mistranslation events involving D-amino acids
Improvement of recombinant protein quality in biotechnology applications
Biosensor development:
Creation of DTD-based biosensors for D-amino acid detection
Environmental monitoring of D-amino acids in marine samples
Potential applications in food safety and fermentation monitoring
Synthetic biology tools:
Integration into synthetic genetic circuits as a quality control component
Development of orthogonal translation systems with enhanced fidelity
Creation of minimal synthetic cells with defined quality control mechanisms
Marine biotechnology applications:
Utilization of R. baltica DTD's potential adaptations to marine conditions
Development of enzyme technologies optimized for high-salt environments
Applications in marine bioremediation or resource utilization
To realize these applications, further characterization of R. baltica DTD's biochemical properties, substrate specificity, and stability under various conditions would be required. The enzyme's potential adaptations to marine environments might provide unique advantages for certain biotechnology applications compared to DTD enzymes from other organisms.