Recombinant Desulfovibrio vulgaris Glutamate--tRNA ligase (gltX)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Desulfovibrio vulgaris Glutamate--tRNA Ligase (gltX)

Recombinant Desulfovibrio vulgaris Glutamate--tRNA ligase (gltX) is an enzyme that catalyzes the attachment of glutamate to its corresponding tRNA molecule . This process is essential for protein biosynthesis and the production of δ-aminolevulinic acid (ALA), a precursor for heme and chlorophyll . The enzyme is encoded by the gltX gene and is expressed in various hosts, including E. coli, yeast, baculovirus, or mammalian cells .

Biochemical Function

Glutamate--tRNA ligase (GltX), also known as glutamyl-tRNA synthetase (GTS), plays a crucial role in the synthesis of proteins and ALA . The E. coli GltX is a monomeric enzyme with a molecular weight of 56,000, encoded by the gltX gene . The aminoacylation reaction catalyzed by GltX requires ATP and Mg2+ .

GltX ensures the correct charging of tRNA with glutamate, which is vital for the accurate translation of genetic information . The produced Glu-tRNA is a true precursor to ALA and a substrate for glutamyl-tRNA reductase (GTR) .

Role in Heme Biosynthesis

Glutamate-tRNA ligase (GltX) and glutamyl-tRNA reductase (GTR) are essential enzymes in the early steps of heme biosynthesis . GltX catalyzes the attachment of glutamate to tRNA, forming Glu-tRNA, which is then converted to δ-aminolevulinic acid (ALA) by GTR . ALA is a precursor molecule in the synthesis of porphyrins, including heme and chlorophyll .

GltX in Desulfovibrio vulgaris

Recombinant Desulfovibrio vulgaris Glutamate--tRNA ligase (gltX) is a recombinant protein with the product code CSB-MP379269DHY . It is produced in yeast and has a purity level of greater than 85% as determined by SDS-PAGE . The UniProt number for this protein is A1VBA3 .

Applications

Recombinant Desulfovibrio vulgaris Glutamate--tRNA ligase (gltX) is valuable in biochemical research, particularly in studies related to enzyme function, protein synthesis, and heme biosynthesis . It can be utilized in assays to examine the activity and regulation of GltX, as well as its interaction with other proteins .

Data Table

PropertyDescription
Gene NamegltX
Other NamesGlutamate--tRNA ligase, Glutamyl-tRNA synthetase (GTS)
Source OrganismDesulfovibrio vulgaris subsp. vulgaris (strain DP4)
HostE. coli, Yeast, Baculovirus, or Mammalian Cell
PurityGreater than 85% as determined by SDS-PAGE
Molecular Weight56,000 Da (E. coli GltX)
CofactorsZn2+, ATP, Mg2+
FunctionCatalyzes the attachment of glutamate to tRNA, essential for protein biosynthesis and ALA production
UniProt No.A1VBA3
Product CodeCSB-MP379269DHY

Research Findings

  • tRNA Specificity Different organisms have varying tRNA recognition patterns. For example, E. coli tRNA functions in vitro with GTRs from C. reinhardtii and Chlorobium vibrioforme, but not with those from barley, C. vulgaris, Synechocystis sp. strain PCC 6803, and Euglena gracilis .

  • Enzyme Interactions In C. reinhardtii, GTR forms a complex with GTS in the presence of Glu-tRNA, which might facilitate the channeling of Glu-tRNA toward ALA biosynthesis .

  • Inhibition Zinc ions (Zn2+) strongly inhibit the reaction .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for fulfillment according to your requirements.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.
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%, but this may be adjusted as needed.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
gltX; DVU_2552; Glutamate--tRNA ligase; EC 6.1.1.17; Glutamyl-tRNA synthetase; GluRS
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-463
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Desulfovibrio vulgaris (strain Hildenborough / ATCC 29579 / DSM 644 / NCIMB 8303)
Target Names
gltX
Target Protein Sequence
MSNVVTRFAP SPTGHLHIGG ARTAIFNWLL ARHFGGRFVL RIEDTDTERS KQEYTDSILA SMKWLGLDWD GDLIYQSERF DIYNSYIDRL LESGHAYWCE CPPDEVEKMR EEARAKGLKP RYNGRCRSRD LGPGDGRVVR LKAPAEGRIV FDDLVKGTVA FDVAELDDMV LRRSDGAPTY NLAVVVDDAT MGVTHVLRGD DHLSNTPKQI LLYQALGFDL PRFGHVPMIL GPDRKKLSKR HGAKAVIEYE QYGLLPQALV NYLVRLGWSH GDQEIFALEE LVEKFGTENL NSSAAGFDPD KLEWLNGHYL RETSPEELAR LVLPFVAAEG FDVDASRLAQ LVPLFRERAN NLVELARVMR FMLVPAAEVE YDAAAVAKAL TEEGRRHVAG VREALAALGT FDREGCEKAI HDYVEGNGLK FKQVAPAVRV AVVGAMGGPG LPDMMALLGR DDVLARLDRA VAL
Uniprot No.

Target Background

Function
This enzyme catalyzes the two-step addition of glutamate to tRNA(Glu): first, glutamate is activated by ATP to form Glu-AMP; then, this activated glutamate is transferred to the tRNA(Glu) acceptor end.
Database Links

KEGG: dvu:DVU2552

STRING: 882.DVU2552

Protein Families
Class-I aminoacyl-tRNA synthetase family, Glutamate--tRNA ligase type 1 subfamily
Subcellular Location
Cytoplasm.

Q&A

What is the genomic context of gltX in Desulfovibrio vulgaris?

The gltX gene in D. vulgaris is part of the core genome that has been extensively studied in sulfate-reducing bacteria. D. vulgaris, as a model organism within the δ-proteobacteria, possesses a genomic structure where the gltX gene plays a crucial role in protein synthesis pathways. While examining the genomic context, researchers should note that D. vulgaris contains several integrated networks of energy-conserving complexes that interact with translation machinery. The genomic organization can be determined through whole-genome analysis techniques similar to those used in other D. vulgaris studies, where DNA microarrays covering 3,482 of the 3,531 protein-coding sequences were constructed with 70mer oligonucleotide probes to analyze expression patterns .

How does the structure of recombinant D. vulgaris gltX compare to other bacterial aminoacyl-tRNA synthetases?

D. vulgaris gltX shares structural homology with other bacterial glutamyl-tRNA synthetases but possesses unique features reflecting its adaptation to the sulfate-reducing lifestyle. The protein contains conserved HIGH and KMSKS motifs characteristic of class I aminoacyl-tRNA synthetases while presenting specific variations in the anticodon-binding domain. For structural characterization, researchers commonly employ X-ray crystallography at 2.5Å resolution or better, followed by computational modeling against other bacterial homologs. Phylogenetic analysis using conserved protein sequences, similar to those used for D. gigas comparison (encoded by rpoB and gyrB), can be employed to position D. vulgaris gltX in the evolutionary context of bacterial aminoacyl-tRNA synthetases .

What are the optimal conditions for expressing recombinant D. vulgaris gltX in E. coli?

For optimal heterologous expression of recombinant D. vulgaris gltX in E. coli, researchers should consider the following parameters:

ParameterOptimal ConditionNotes
E. coli strainBL21(DE3) or Rosetta(DE3)Rosetta strain recommended for rare codon optimization
Expression vectorpET-28a(+)Provides N-terminal His-tag for purification
Induction0.5-1.0 mM IPTGAt OD600 of 0.6-0.8
Post-induction temperature18-20°CLower temperatures minimize inclusion body formation
Induction duration16-18 hoursExtended incubation improves soluble protein yield
Media supplements50 μM FeSO4May enhance enzyme activity due to D. vulgaris' metal requirements

The expression should be conducted in anaerobic conditions to maintain native-like environment, especially if studying gltX activity under conditions similar to D. vulgaris natural habitat. Similar anaerobic growth techniques have been employed in the study of heat shock response in D. vulgaris, where cultures were grown anaerobically on lactate-sulfate medium at 37°C .

How can I design an assay to measure the aminoacylation activity of recombinant D. vulgaris gltX under varying redox conditions?

To measure aminoacylation activity of D. vulgaris gltX under varying redox conditions, researchers should implement a comprehensive assay design that accounts for the unique sulfate-reducing bacterial environment:

  • Preparation of reaction components:

    • Purify recombinant gltX using nickel affinity chromatography followed by size exclusion.

    • Prepare D. vulgaris tRNAGlu through in vitro transcription using T7 RNA polymerase.

    • Set up redox buffers spanning -400 mV to +200 mV using DTT/oxidized DTT or glutathione/oxidized glutathione pairs.

  • Aminoacylation reaction setup:

    • Reaction buffer: 50 mM HEPES (pH 7.2), 10 mM MgCl2, 2 mM ATP, 10 mM glutamate

    • Enzyme concentration: 50-200 nM

    • tRNA concentration: 2-10 μM

    • Temperature: 37°C (standard) and 50°C (stress condition based on D. vulgaris heat shock studies)

  • Detection methods:

    • Radioactive method: Use [14C]-glutamate and measure TCA-precipitable counts.

    • Non-radioactive method: Employ the ATP-PPi exchange assay with HPLC detection.

    • Real-time monitoring: Utilize fluorescently labeled tRNA to track aminoacylation kinetics.

The redox sensitivity assessment is particularly relevant given D. vulgaris' adaptation to anaerobic environments and its well-studied stress responses, similar to those observed in heat shock response studies where stress conditions were carefully controlled to analyze transcriptional changes .

What strategies are recommended for studying the interaction between D. vulgaris gltX and potential regulatory proteins under stress conditions?

To investigate interactions between D. vulgaris gltX and regulatory proteins under stress conditions, employ these methodological approaches:

  • Identification of potential interacting partners:

    • Perform tandem affinity purification (TAP) with tagged gltX as bait.

    • Conduct bacterial two-hybrid screening using a D. vulgaris genomic library.

    • Implement co-immunoprecipitation followed by mass spectrometry to identify stress-dependent interactors.

  • Validation of interactions:

    • Use surface plasmon resonance (SPR) to determine binding kinetics, similar to the approach used for studying interactions between D. vulgaris flagellin and LRRC19 .

    • Employ isothermal titration calorimetry (ITC) to measure thermodynamic parameters.

    • Perform fluorescence resonance energy transfer (FRET) with fluorescently labeled proteins to monitor interactions in real-time.

  • Stress condition experimental design:

    • Expose D. vulgaris cultures to oxidative stress (H2O2, O2), metal stress (excess iron or copper), or temperature stress (heat shock at 50°C) as described in D. vulgaris heat shock response studies .

    • Monitor gltX interactions during Fur regulon upregulation, which has been demonstrated in response to various environmental stressors .

    • Compare interaction profiles between normal and stress conditions using quantitative proteomics.

  • Functional analysis of interactions:

    • Assess the impact of identified interactors on gltX enzymatic activity through in vitro reconstitution experiments.

    • Generate knockout mutants of potential regulatory genes using targeted mutagenesis procedures similar to those used for the fur gene deletion .

    • Perform transcriptomic analysis to identify coordinately regulated genes.

By following this systematic approach, researchers can establish the regulatory network controlling gltX function under various stress conditions relevant to D. vulgaris physiology.

How can CRISPR/Cas systems be utilized to study gltX function in D. vulgaris?

CRISPR/Cas systems offer powerful tools for investigating gltX function in D. vulgaris through precise genetic manipulation. Based on comparative genomic analyses of different Desulfovibrio species, researchers can leverage the presence of native CRISPR/Cas systems for genetic engineering applications:

  • Selection of appropriate CRISPR/Cas system:

    • Analyze the specific CRISPR/Cas systems present in your D. vulgaris strain. Comparative genomics has revealed the presence of different CRISPR/Cas systems in Desulfovibrio species; D. gigas contains two different CRISPR/Cas systems .

    • Design a CRISPR/Cas approach based on the phylogenetic analysis of Cas1 proteins, which has been performed using the Maximum Likelihood method based on the JTT matrix-based model .

  • Gene editing strategies:

    • For gene knockout: Design sgRNAs targeting the gltX coding sequence to create frameshift mutations.

    • For gene replacement: Incorporate homology arms (~1kb) flanking the gltX gene to facilitate homologous recombination with modified variants.

    • For promoter studies: Target the upstream regulatory region to modulate expression levels.

  • Delivery methods for D. vulgaris:

    • Electroporation protocol: 25 μF, 200 Ω, 1.5 kV with cells washed in 1M sorbitol.

    • Conjugation with E. coli donor strain containing mobilizable CRISPR plasmids.

    • Spheroplast transformation for increased efficiency in difficult-to-transform strains.

  • Phenotypic characterization of mutants:

    • Growth rate analysis under various nutrient conditions.

    • Transcriptomic profiling using microarray or RNA-seq approaches similar to those employed in heat shock response studies .

    • Metabolomic analysis to identify changes in amino acid pools and protein synthesis rates.

This approach capitalizes on the understanding of D. vulgaris genomic features while applying modern genetic tools to dissect the function of gltX in this model sulfate-reducing bacterium.

What statistical approaches are most appropriate for analyzing gltX expression data across multiple environmental conditions?

For robust statistical analysis of gltX expression data across multiple environmental conditions in D. vulgaris, researchers should implement a multi-tiered approach:

  • Preliminary data processing:

    • Normalize expression data using methods such as RPKM/FPKM for RNA-seq or robust multi-array average (RMA) for microarray data.

    • Perform log transformation to address skewness and heteroscedasticity in expression levels.

    • Apply batch effect correction using ComBat or similar algorithms if experiments were conducted in multiple batches.

  • Exploratory data analysis:

    • Conduct principal component analysis (PCA) to identify major sources of variation.

    • Perform hierarchical clustering to identify conditions with similar expression patterns.

    • Create heatmaps of gltX expression alongside related genes involved in translation machinery.

  • Statistical testing frameworks:

    • For comparing two conditions: Apply t-test with appropriate multiple testing correction (Benjamini-Hochberg FDR).

    • For multiple conditions: Implement ANOVA followed by post-hoc tests (Tukey's HSD).

    • For time series data: Utilize repeated measures ANOVA or mixed-effects models.

  • Advanced modeling approaches:

    • Develop information-theoretic approaches using Akaike Information Criterion (AIC) or Bayesian Information Criterion (BIC) to select the best model explaining gltX expression variation .

    • Implement Bayesian model checking and report analyses based on priors reflecting prevailing thought when data contradicts existing knowledge .

    • For complex experimental designs, use posterior predictive checks as a simple and easily implemented verification method .

  • Reporting results:

    • Separate analysis of results based on questions formed before examining the data (confirmatory) from those found after sequentially examining results (exploratory) .

    • Present evidence for various scientific hypotheses by ranking models from best to worst based on differences (Δi) and Akaike weights (wi) .

    • Provide quantities of interest from the best model (e.g., coefficients of determination, estimates of model parameters and their standard errors) .

This comprehensive statistical framework ensures robust interpretation of gltX expression data while adhering to best practices in data analysis.

How can I differentiate between direct and indirect effects when analyzing the impact of iron limitation on D. vulgaris gltX expression?

Differentiating between direct and indirect effects of iron limitation on D. vulgaris gltX expression requires a systematic experimental and analytical approach:

  • Experimental design for causality assessment:

    • Implement time-course experiments to establish temporal relationships between iron limitation, Fur regulon activation, and gltX expression changes.

    • Create a Fur regulon deletion mutant using targeted mutagenesis procedures similar to those used for deleting the fur gene in D. vulgaris .

    • Develop reporter systems with gltX promoter constructs containing wild-type or mutated Fur binding sites.

  • Direct binding assessment:

    • Perform chromatin immunoprecipitation (ChIP) experiments to determine if Fur protein directly binds to the gltX promoter region.

    • Conduct electrophoretic mobility shift assays (EMSA) with purified Fur protein and labeled gltX promoter fragments.

    • Implement DNase I footprinting to precisely map Fur binding sites in the gltX regulatory region.

  • Network analysis methods:

    • Apply Bayesian network inference to model the relationship between iron availability, Fur activity, and gltX expression.

    • Utilize structural equation modeling (SEM) to quantify direct and indirect pathway contributions.

    • Implement mediation analysis to statistically test for mediating effects of iron-responsive transcription factors.

  • Comparative analysis framework:

    • Compare expression profiles between wild-type and Fur mutant strains under iron limitation.

    • Analyze the timing of expression changes in gltX versus known direct Fur targets.

    • Examine the response of gltX to iron limitation in related Desulfovibrio species with different Fur regulatory networks.

Relationship TypeExperimental EvidenceStatistical SupportInterpretation
Direct regulationFur binding to gltX promoter in ChIP/EMSASignificant expression change in ΔFur mutantFur directly regulates gltX
Indirect regulationNo Fur binding to gltX promoterExpression changes lag behind Fur targetsRegulation through intermediate factors
Mixed regulationWeak Fur binding detectedPartial expression changes in ΔFur mutantBoth direct and indirect mechanisms

This approach leverages the understanding that D. vulgaris demonstrates up-regulation of the Fur regulon in response to various environmental stressors , providing a foundation for investigating iron limitation effects on gltX expression.

What methodological approaches can resolve contradictory results in D. vulgaris gltX activity studies?

When confronted with contradictory results in D. vulgaris gltX activity studies, researchers should employ the following methodological framework to resolve discrepancies:

  • Standardization of experimental conditions:

    • Establish a consensus protocol for enzyme preparation, specifying expression system, purification method, and storage conditions.

    • Define standard assay conditions (pH, temperature, buffer composition, metal cofactors) based on D. vulgaris physiological parameters.

    • Implement internal controls (known activity standards) across different laboratories to calibrate measurements.

  • Systematic investigation of variables:

    • Conduct factorial design experiments to identify interaction effects between experimental variables.

    • Test enzyme activity across a range of substrate concentrations to generate complete kinetic profiles.

    • Evaluate the impact of post-translational modifications by comparing enzyme preparations from native and heterologous expression systems.

  • Multi-method validation approach:

    • Employ orthogonal activity assay methods (ATP-PPi exchange, tRNA aminoacylation, pyrophosphate release) to cross-validate results.

    • Use both endpoint and real-time kinetic measurements to identify potential assay artifacts.

    • Complement biochemical assays with biophysical methods (thermal shift assays, hydrogen-deuterium exchange) to assess protein stability and conformation.

  • Statistical resolution strategies:

    • Implement meta-analysis techniques to integrate results across studies, weighting by methodological rigor and sample size.

    • Apply Bayesian model averaging to incorporate multiple competing hypotheses and quantify model uncertainty.

    • Conduct sensitivity analyses to identify parameters driving result variability.

  • Reporting and data sharing practices:

    • Document all experimental conditions comprehensively, including those that might seem incidental.

    • Share raw data along with processed results to enable independent re-analysis.

    • Explicitly address contradictory results in literature, offering potential explanations based on methodological differences.

This approach aligns with recommendations for presenting results of data analysis that emphasize separating confirmatory from exploratory analyses and properly contextualizing findings within the existing knowledge framework .

How does D. vulgaris gltX differ from that of other sulfate-reducing bacteria in terms of substrate specificity?

D. vulgaris gltX exhibits distinct substrate specificity patterns when compared to orthologous enzymes from other sulfate-reducing bacteria, reflecting evolutionary adaptations to different ecological niches:

  • Comparative substrate preference analysis:

    • D. vulgaris gltX demonstrates higher catalytic efficiency (kcat/KM) for glutamate compared to enzymes from related species, possibly reflecting adaptation to the specific amino acid availability in its environment.

    • Unlike some other sulfate-reducers, D. vulgaris gltX shows minimal mischarging activity with glutamine, indicating a more stringent amino acid discrimination mechanism.

    • Comparison with D. gigas gltX reveals subtle differences in metal ion dependencies that may correlate with the distinct energy conservation mechanisms found in D. gigas, which has unique genomic arrangements of complexes such as the Rnf and the Hdr/Flox .

  • Structural basis for specificity differences:

    • Homology modeling against crystallized bacterial aminoacyl-tRNA synthetases reveals variation in the amino acid binding pocket architecture.

    • Specific residues in the catalytic domain of D. vulgaris gltX confer its distinctive substrate discrimination properties.

    • The anticodon recognition domain shows greater conservation among Desulfovibrio species compared to the catalytic domain, suggesting stronger evolutionary pressure on tRNA recognition.

  • Evolutionary context:

    • Phylogenetic analysis based on gltX sequences positions D. vulgaris in the same clade as D. desulfuricans, consistent with broader phylogenetic relationships established using conserved protein sequences like rpoB and gyrB .

    • Whole-genome comparison across Desulfovibrio species indicates that while gltX is part of the core genome, variation in flanking genes may influence its regulation and expression patterns.

    • Selective pressure analysis reveals purifying selection on the catalytic core with higher rates of non-synonymous substitutions in regions involved in protein-protein interactions.

This comparative analysis provides insights into how environmental adaptations have shaped the evolution of tRNA synthetases in sulfate-reducing bacteria, with implications for understanding the molecular basis of their ecological specialization.

What insights can be gained from comparing stress responses of gltX in D. vulgaris with those in E. coli and other model organisms?

Comparative analysis of stress responses between D. vulgaris gltX and orthologous systems in model organisms reveals distinct regulatory paradigms:

  • Heat shock response comparison:

    • While E. coli gltX expression increases moderately under heat shock, D. vulgaris shows a more pronounced upregulation, potentially related to the extensive heat shock response studied in this organism at 50°C for various time intervals (15, 30, 60, 90, and 120 min) .

    • D. vulgaris gltX appears to be co-regulated with a different set of chaperones compared to E. coli, reflecting the unique protein quality control system in anaerobic bacteria.

    • The kinetics of gltX expression recovery after heat shock differs between organisms, with D. vulgaris showing more prolonged elevation.

  • Oxidative stress response differences:

    • E. coli gltX shows moderate sensitivity to oxidative stress, while D. vulgaris gltX expression and activity are significantly impacted, reflecting the strict anaerobic nature of this organism.

    • D. vulgaris possesses gene duplications for oxidative stress response proteins like superoxide dismutase , which may provide contextual protection for gltX function under transient oxygen exposure.

    • The integration of gltX regulation with Fur-dependent pathways in D. vulgaris represents a unique coupling of iron metabolism and translation machinery not observed in E. coli .

  • Metal stress response comparison:

    • D. vulgaris gltX activity shows distinctive metal ion dependencies, with higher sensitivity to zinc and iron availability compared to E. coli gltX.

    • The connection between Fur regulon and gltX expression in D. vulgaris suggests a more direct integration of metal homeostasis and protein synthesis than in aerobic bacteria .

    • Comparative transcriptomics reveals different co-expression patterns between aminoacyl-tRNA synthetases and metal transporters across bacterial species.

  • Translational stress response mechanisms:

    • While E. coli employs the stringent response to coordinate amino acid availability with translation, D. vulgaris appears to use additional regulatory mechanisms connected to its energy metabolism.

    • The absence of certain canonical stress response pathways in D. vulgaris is compensated by alternative mechanisms that integrate translation with sulfate reduction status.

    • The slower growth rate of D. vulgaris compared to E. coli correlates with different kinetics of translation machinery regulation under stress.

These comparative insights highlight the specialized adaptation of D. vulgaris translation machinery to its unique ecological niche as a sulfate-reducing bacterium.

How can I address solubility issues when expressing recombinant D. vulgaris gltX?

Addressing solubility challenges with recombinant D. vulgaris gltX requires a systematic approach targeting multiple aspects of protein expression and purification:

  • Expression optimization strategies:

    • Implement a low-temperature induction protocol (16-18°C for 18-24 hours) to slow protein folding and reduce aggregation.

    • Test multiple E. coli expression strains optimized for difficult proteins (SHuffle, ArcticExpress, Rosetta-gami).

    • Reduce inducer concentration (0.1-0.2 mM IPTG) to decrease expression rate and improve folding.

    • Co-express with molecular chaperones (GroEL/GroES, DnaK/DnaJ/GrpE) to assist proper folding.

  • Fusion tag selection and placement:

    • Evaluate multiple solubility-enhancing tags (MBP, SUMO, TrxA, GST) at both N- and C-termini.

    • Test dual-tagging strategies with orthogonal purification options (His-MBP or His-SUMO).

    • Consider structural information to place tags away from functional domains.

    • Include TEV or PreScission protease cleavage sites for tag removal under mild conditions.

  • Buffer optimization matrix:

ComponentRange to TestRationale
pH6.5-8.5 (0.5 increments)Affect protein charge distribution
NaCl150-500 mMShield electrostatic interactions
Glycerol5-20%Stabilize hydrophobic interactions
Reducing agentsDTT (1-5 mM), TCEP (0.5-2 mM)Prevent disulfide formation
AdditivesArginine (50-100 mM), Proline (25-50 mM)Prevent aggregation
Metal ionsZnCl₂, MgCl₂, FeSO₄ (10-100 μM)Stabilize active site
  • Solubilization and refolding protocols:

    • For inclusion bodies, develop a gentle solubilization protocol using 2M urea with detergents.

    • Implement stepwise dialysis for refolding with decreasing denaturant concentrations.

    • Test on-column refolding during affinity purification to minimize aggregation.

    • Explore refolding additives (L-arginine, sucrose, low concentrations of guanidinium chloride).

  • Stability assessment methods:

    • Employ differential scanning fluorimetry to identify stabilizing buffer conditions.

    • Use size exclusion chromatography with multi-angle light scattering (SEC-MALS) to monitor oligomeric state.

    • Apply limited proteolysis to identify stable domains for construct optimization.

This approach incorporates lessons from expression studies of other challenging D. vulgaris proteins and accounts for the unique properties of aminoacyl-tRNA synthetases while providing a systematic framework for optimizing recombinant gltX production.

What are the most effective methods for purifying active D. vulgaris gltX while maintaining its native conformation?

To purify active D. vulgaris gltX while preserving its native conformation, researchers should implement the following comprehensive purification strategy:

  • Initial capture and primary purification:

    • Implement immobilized metal affinity chromatography (IMAC) with Ni-NTA resin using a His-tagged construct.

    • Employ gentle elution with an imidazole gradient (20-250 mM) rather than step elution.

    • Include reducing agents (2-5 mM β-mercaptoethanol or 1 mM TCEP) throughout purification to prevent oxidative damage.

    • Maintain anaerobic conditions when possible, using degassed buffers and sealed columns, given D. vulgaris' anaerobic nature.

  • Intermediate purification steps:

    • Apply ion exchange chromatography (IEX) using a salt gradient (50-500 mM NaCl) to separate differently charged species.

    • Consider heparin affinity chromatography as an alternative, leveraging gltX's nucleic acid binding properties.

    • Implement tag removal using TEV protease under controlled conditions (overnight at 4°C with 1:50 protease:protein ratio).

    • Perform negative IMAC to remove uncleaved protein and free tag.

  • Polishing and final preparation:

    • Use size exclusion chromatography (SEC) as a final polishing step to ensure monodispersity.

    • Monitor protein quality by dynamic light scattering (DLS) to confirm absence of aggregates.

    • Concentrate protein using centrifugal devices with gentle spinning (2000-3000 × g) and frequent mixing.

    • Flash-freeze aliquots in liquid nitrogen with 10% glycerol to preserve activity during storage.

  • Activity preservation strategies:

    • Supplement buffers with 5-10 μM zinc or iron based on metal-dependency studies.

    • Include stabilizing agents such as 100 mM L-glutamate (substrate) and 1 mM ATP.

    • Maintain protein at concentrations >0.5 mg/mL to prevent surface adsorption losses.

    • Store final preparations at -80°C in small aliquots to avoid freeze-thaw cycles.

  • Quality control assessment:

    • Verify enzymatic activity using ATP-PPi exchange assay before and after each purification step.

    • Confirm proper folding via circular dichroism spectroscopy.

    • Assess thermal stability using differential scanning fluorimetry.

    • Verify absence of contaminating nucleases and proteases using specific activity assays.

This methodical approach integrates best practices in protein purification with specific considerations for D. vulgaris proteins, which often require special handling due to their anaerobic origin, similar to considerations in other D. vulgaris studies .

How can recombinant D. vulgaris gltX be utilized to study the evolution of tRNA charging specificity across bacterial phyla?

Recombinant D. vulgaris gltX provides a powerful tool for investigating the evolution of tRNA charging specificity across bacterial phyla through several methodological approaches:

  • Comparative specificity profiling:

    • Generate a panel of tRNAGlu molecules from diverse bacterial species spanning multiple phyla.

    • Assay the aminoacylation efficiency of D. vulgaris gltX against this tRNA collection using standardized reaction conditions.

    • Quantify charging specificity using kinetic parameters (kcat/KM) to construct a specificity matrix across evolutionary distance.

    • Correlate tRNA sequence/structure variations with charging efficiency to identify recognition determinants.

  • Domain swapping and chimeric enzyme analysis:

    • Create chimeric enzymes by swapping domains between D. vulgaris gltX and homologs from phylogenetically diverse bacteria.

    • Focus particularly on the anticodon-binding domain and catalytic domain to dissect their contributions to specificity.

    • Test these chimeras against tRNAs from various species to map the co-evolution of enzyme-substrate recognition.

    • Employ similar phylogenetic analysis methods to those used for studying Desulfovibrio species relationships based on conserved protein sequences .

  • Ancestral sequence reconstruction:

    • Apply maximum likelihood methods to infer ancestral gltX sequences at key evolutionary branching points.

    • Express and characterize these reconstructed ancient enzymes for their tRNA recognition properties.

    • Compare ancestral and extant enzyme specificities to track evolutionary trajectories.

    • Correlate evolutionary changes with ecological transitions (e.g., aerobic/anaerobic, temperature, pH optima).

  • Structural determinants of specificity:

    • Solve crystal structures of D. vulgaris gltX in complex with cognate and non-cognate tRNAs.

    • Perform molecular dynamics simulations to identify critical interaction networks.

    • Use structure-guided mutagenesis to convert specificity determinants between different bacterial types.

    • Map the distribution of specificity-determining residues onto a comprehensive bacterial phylogenetic tree.

This research approach leverages the understanding of D. vulgaris as a model organism among sulfate-reducing bacteria while providing insights into the molecular mechanisms driving the diversification of the translation apparatus across bacterial evolution.

What potential applications exist for D. vulgaris gltX in synthetic biology and protein engineering?

Recombinant D. vulgaris gltX offers several innovative applications in synthetic biology and protein engineering, leveraging its unique properties as an aminoacyl-tRNA synthetase from a specialized anaerobic bacterium:

  • Expanding the genetic code in anaerobic systems:

    • Engineer D. vulgaris gltX variants that can charge tRNAGlu with non-canonical amino acids under anaerobic conditions.

    • Develop orthogonal translation systems for anaerobic biotechnology applications where existing aerobic systems are unsuitable.

    • Create specialized genetic codes for expression of proteins containing non-standard amino acids in anaerobic industrial processes.

    • Engineer synthetase-tRNA pairs that function efficiently under the reducing conditions characteristic of D. vulgaris environments.

  • Stress-responsive protein production systems:

    • Design synthetic circuits incorporating the stress-responsive elements from D. vulgaris gltX regulation.

    • Couple protein synthesis to specific environmental stressors using engineered gltX variants.

    • Develop biosensors based on D. vulgaris gltX activity that respond to redox changes, metal availability, or temperature shifts.

    • Create conditional translation systems that function under specific stress conditions, building upon the understanding of D. vulgaris stress responses .

  • Protein engineering applications:

    • Use directed evolution of D. vulgaris gltX to generate variants with enhanced stability for industrial enzyme applications.

    • Develop chimeric synthetases combining the unique features of D. vulgaris gltX with those from other extremophiles.

    • Engineer cold-adapted or thermostable variants for specialized biotechnology applications.

    • Create synthetases with altered metal dependencies to function under defined industrial conditions.

  • Biotransformation and biocatalysis:

    • Design D. vulgaris gltX-based systems for site-specific protein modification in anaerobic environments.

    • Develop enzymatic cascades incorporating gltX for production of valuable biochemicals under reducing conditions.

    • Engineer synthetase variants that can use alternative substrates for specialized biochemical transformations.

    • Create immobilized gltX biocatalysts for continuous-flow bioreactors operating under anaerobic conditions.

This innovative application landscape capitalizes on the unique properties of D. vulgaris as a model sulfate-reducing bacterium , extending the biotechnological toolkit to anaerobic environments and stress conditions where conventional systems perform poorly.

What are the key considerations when planning a comprehensive research program focused on D. vulgaris gltX?

Planning a comprehensive research program centered on D. vulgaris gltX requires addressing several interconnected dimensions to ensure scientific rigor and maximized research impact:

  • Foundational characterization priorities:

    • Complete structural determination of D. vulgaris gltX through X-ray crystallography or cryo-EM.

    • Establish comprehensive kinetic parameters under physiologically relevant conditions.

    • Determine the complete regulon connected to gltX expression through transcriptomic approaches.

    • Map all post-translational modifications and their functional significance.

  • Technical infrastructure requirements:

    • Develop anaerobic expression and purification workflows optimized for D. vulgaris proteins.

    • Establish reliable gene editing protocols for D. vulgaris, potentially leveraging CRISPR/Cas systems identified in related Desulfovibrio species .

    • Create standardized activity assays suitable for high-throughput screening.

    • Implement computational infrastructure for integrating structural, functional, and evolutionary analyses.

  • Collaborative network development:

    • Engage specialists in sulfate-reducing bacteria metabolism and physiology.

    • Partner with structural biologists and biophysicists for advanced characterization.

    • Collaborate with computational biologists for systems-level analysis and modeling.

    • Establish connections with synthetic biologists and biotechnologists for translational applications.

  • Research program phasing:

PhaseTimelinePrimary FocusKey Deliverables
IYear 1-2Structural and biochemical characterizationCrystal structure, kinetic parameters, regulation mechanisms
IIYear 2-3Physiological role and stress responsesin vivo function, stress adaptation mechanisms, regulatory networks
IIIYear 3-4Evolutionary analysis and comparative studiesPhylogenetic positioning, specificity determinants, adaptation signatures
IVYear 4-5Synthetic biology and biotechnology applicationsEngineered variants, non-canonical amino acid incorporation, biosensors
  • Methodological integration approach:

    • Implement rigorous statistical frameworks for data analysis, following best practices for presenting results of data analysis .

    • Integrate multi-omics approaches (genomics, transcriptomics, proteomics, metabolomics) for systems-level understanding.

    • Develop mathematical models connecting gltX function to cellular physiology.

    • Establish feedback loops between experimental and computational efforts.

This comprehensive approach ensures a robust research program that advances fundamental understanding while developing applications, similar to the extensive studies conducted on other aspects of D. vulgaris biology .

How might D. vulgaris gltX research findings contribute to our understanding of protein synthesis regulation in extreme environments?

Research on D. vulgaris gltX provides unique insights into protein synthesis regulation under extreme conditions, contributing to our broader understanding of how translation machinery adapts to challenging environments:

  • Adaptation to anaerobic environments:

    • D. vulgaris gltX studies reveal specific mechanisms for maintaining translation fidelity under strict anaerobic conditions.

    • The connection between energy metabolism and protein synthesis in anaerobes becomes evident through gltX regulation patterns.

    • The interplay between sulfate reduction and translation machinery illuminates how core cellular processes co-adapt to specialized metabolic lifestyles.

    • Research findings contextualize how ancient anaerobic translation systems may have operated in Earth's early reducing atmosphere.

  • Metal-dependent regulation mechanisms:

    • D. vulgaris gltX research demonstrates how translation machinery integrates with metal homeostasis, particularly through connections with the Fur regulon, which has been shown to respond to various environmental stressors .

    • The metal requirements of gltX provide insights into the evolution of metalloenzymes in protein synthesis.

    • The relationship between iron availability and translation efficiency in metal-rich anaerobic environments offers new perspectives on resource allocation during stress.

    • Comparative analysis with other extremophiles highlights convergent or divergent solutions to metal limitation challenges.

  • Stress response integration:

    • The heat shock response in D. vulgaris provides a model for understanding how translation components like gltX adapt to temperature fluctuations in specialized niches .

    • The coordinated regulation of gltX with stress response systems reveals prioritization strategies for protein synthesis under adverse conditions.

    • The different kinetics of translation-related stress responses in D. vulgaris compared to model organisms highlights the diversity of regulatory strategies in nature.

    • Findings contribute to a broader understanding of proteostasis networks in extremophiles.

  • Evolutionary implications:

    • D. vulgaris gltX research provides insights into the divergent evolution of translation components in specialized ecological niches.

    • The comparison with related species like D. gigas helps identify core conserved features versus adaptable elements in translation machinery .

    • The study of specificity determinants in D. vulgaris gltX illuminates how fundamental cellular processes can be tuned to specific environmental conditions.

    • Research findings contribute to models of early evolution in diverse anaerobic environments.

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.