Recombinant Methanococcus maripaludis Glutamyl-tRNA (Gln) amidotransferase subunit E (gatE), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing 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. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type is determined during production. Please specify your desired tag type for preferential development.
Synonyms
gatE; MMP1265Glutamyl-tRNA(Gln) amidotransferase subunit E; Glu-ADT subunit E; EC 6.3.5.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Methanococcus maripaludis (strain S2 / LL)
Target Names
gatE
Uniprot No.

Target Background

Function
This protein facilitates the formation of correctly charged Gln-tRNA(Gln) by transamidating misacylated Glu-tRNA(Gln) in organisms lacking glutaminyl-tRNA synthetase. This reaction occurs in the presence of glutamine and ATP, utilizing an activated gamma-phospho-Glu-tRNA(Gln) intermediate. The GatDE system exhibits specificity for glutamate and does not act on aspartate.
Database Links

KEGG: mmp:MMP1265

STRING: 267377.MMP1265

Protein Families
GatB/GatE family, GatE subfamily

Q&A

What is Methanococcus maripaludis and why is it significant as a model organism?

Methanococcus maripaludis is a mesophilic, hydrogenotrophic methanogen belonging to the kingdom Euryarchaeota in the domain Archaea. This organism possesses a single circular chromosome of 1,661,137 bp containing 1,722 protein-coding genes, with a relatively low G+C content of 33.1% . M. maripaludis has emerged as a particularly valuable model organism for several reasons. First, unlike many other methanogens, it is genetically tractable, making it amenable to genetic manipulation and functional studies . Second, it has been extensively characterized at the physiological and molecular levels, with numerous studies employing genetic tools to understand its metabolism and regulation . Finally, while M. maripaludis shares evolutionary relationships with Methanocaldococcus jannaschii (approximately 64% of its ORFs have their highest Blastp hits in M. jannaschii), it possesses many unique features, with about one-third of its genes lacking orthologs in M. jannaschii . These characteristics make M. maripaludis an ideal platform for investigating fundamental archaeal processes, including the unique mechanisms of tRNA charging found in this domain of life.

What is the specific function of the Glutamyl-tRNA (Gln) amidotransferase subunit E (gatE) in archaeal systems?

The Glutamyl-tRNA (Gln) amidotransferase subunit E (gatE) in Methanococcus maripaludis functions as an essential component of the GatDE complex that catalyzes the formation of correctly charged Gln-tRNA(Gln) through a transamidation pathway . This process is particularly important in organisms like M. maripaludis that lack a dedicated glutaminyl-tRNA synthetase. The reaction occurs in multiple steps:

  • First, Glu-tRNA(Gln) is misacylated by a non-discriminating glutamyl-tRNA synthetase

  • The GatDE complex then recognizes this misacylated tRNA

  • In the presence of glutamine and ATP, the complex catalyzes the conversion of the glutamate attached to the tRNA into glutamine via an activated gamma-phospho-Glu-tRNA(Gln) intermediate

Importantly, the GatDE system in M. maripaludis demonstrates substrate specificity, acting exclusively on glutamate and not on aspartate . The gatE subunit works in concert with gatD to form a functional amidotransferase, with gatE likely contributing to tRNA recognition and binding specificity. This indirect pathway for Gln-tRNA(Gln) formation represents an ancient mechanism for accurate translation that differs fundamentally from the direct aminoacylation pathways common in most bacteria and eukaryotes.

How does the archaeal GatDE system differ from bacterial transamidation pathways for tRNA charging?

The archaeal GatDE system found in Methanococcus maripaludis represents a distinct evolutionary solution to the challenge of accurate tRNA charging compared to bacterial systems. Key differences include:

FeatureArchaeal GatDE SystemBacterial GatCAB System
Subunit compositionTwo subunits (gatD and gatE) Three subunits (gatC, gatA, and gatB)
Substrate specificitySpecific for Glu-tRNA(Gln) only Can act on both Glu-tRNA(Gln) and Asp-tRNA(Asn)
Evolutionary originArchaeal-specific innovationWider distribution across bacterial lineages
Structural featuresgatE shows distinct structural elementsDifferent quaternary structure organization
ATP utilizationRequired for the activation step via gamma-phospho-Glu-tRNA(Gln) Similar ATP-dependent mechanism but with distinct catalytic residues

The existence of these different systems highlights the diversity of solutions that have evolved to address the challenge of accurate translation across domains of life. In M. maripaludis, the GatDE system works specifically with glutamate and does not act on aspartate, whereas many bacterial systems show broader substrate specificity . This specialization likely reflects the unique evolutionary pressures and metabolic constraints in archaeal systems.

What expression systems are most effective for producing functional recombinant M. maripaludis gatE?

Producing functional recombinant M. maripaludis gatE requires careful consideration of expression systems that can accommodate the unique properties of archaeal proteins. Based on general principles of recombinant protein production and the specific characteristics of M. maripaludis, the following approaches are recommended:

  • Expression host selection: E. coli BL21(DE3) derivatives typically provide good expression levels, though codon optimization may be necessary given the low G+C content (33.1%) of M. maripaludis genes . For improved protein folding, consider cold-adapted strains like Arctic Express or chaperon-enhanced systems like Rosetta-gami.

  • Vector design considerations:

    • Include a C-terminal His-tag rather than N-terminal to minimize interference with catalytic function

    • Employ inducible promoters like T7 with fine control of expression rates

    • Consider fusion partners like SUMO or thioredoxin to enhance solubility

  • Expression conditions optimization: Using Design of Experiments (DoE) approaches allows systematic evaluation of multiple factors affecting recombinant protein expression . A fractional factorial design examining the following variables often yields optimal conditions:

FactorLow LevelHigh Level
IPTG concentration0.1 mM1 mM
Temperature post-induction16°C30°C
Induction time4 hours18 hours
Media compositionLBDefined minimal
Cell density at inductionOD600 0.4OD600 0.8

The DoE approach provides significant advantages over the inefficient one-factor-at-a-time optimization, as it accounts for interactive effects between variables and reduces experimental time and cost . This approach is particularly valuable given the complex interactions among reagents that make it impossible for one set of reaction conditions to be optimal for all proteins .

How can researchers optimize purification protocols for recombinant gatE while maintaining functional activity?

Purification of recombinant M. maripaludis gatE requires balancing high yield with preservation of functional activity. A methodological approach should consider:

  • Initial extraction optimization:

    • Buffer composition: Tris-HCl (pH 8.0) with 300-500 mM NaCl generally provides good stability

    • Include 5-10% glycerol and 1-5 mM β-mercaptoethanol to maintain protein stability

    • Gentle cell disruption methods (e.g., sonication with cooling intervals) to prevent protein aggregation

  • Multi-step purification strategy:

    • Initial IMAC (Immobilized Metal Affinity Chromatography) using Ni-NTA for His-tagged gatE

    • Secondary purification via ion exchange chromatography (typically anion exchange)

    • Final polishing step using size exclusion chromatography to ensure homogeneity

  • Activity preservation measures:

    • Maintain samples at 4°C throughout purification

    • Include glutamine (substrate) at low concentrations (0.1-1 mM) in purification buffers

    • Test multiple elution conditions using DoE approaches to maximize recovery of active protein

  • Quality assessment metrics:

    • SDS-PAGE for purity evaluation (aim for >95% homogeneity)

    • Western blotting to confirm identity

    • Dynamic light scattering to assess aggregation state

    • Preliminary activity assays at multiple stages to track functional preservation

Importantly, researchers should implement DoE approaches to systematically evaluate how different purification parameters interact to affect final protein activity . This can significantly reduce the time and resources required to develop an optimal purification protocol compared to traditional one-factor-at-a-time optimization approaches.

What assay systems can reliably measure the enzymatic activity of recombinant gatE?

Measuring the enzymatic activity of recombinant gatE from M. maripaludis requires careful consideration of its biological function as part of the GatDE complex that facilitates the transamidation of misacylated Glu-tRNA(Gln) to Gln-tRNA(Gln) . Effective assay approaches include:

  • Coupled spectrophotometric assays:

    • Monitor ATP hydrolysis during the transamidation reaction through coupled enzyme systems

    • Key readout: NADH oxidation (decrease in absorbance at 340 nm)

    • Advantages: Continuous monitoring, quantitative kinetic parameters

    • Limitations: Potential interference from coupling enzymes

  • Thin-layer chromatography (TLC) with radioactive substrates:

    • Use of [14C]-Glu-tRNA(Gln) as substrate and monitoring conversion to [14C]-Gln-tRNA(Gln)

    • Key readout: Separation and quantification of substrate and product spots

    • Advantages: Direct measurement of actual transamidation activity

    • Limitations: Requires radioactive materials, discontinuous measurement

  • Mass spectrometry-based assays:

    • Direct measurement of tRNA aminoacylation status

    • Key readout: Mass shift corresponding to glutamine vs. glutamate attachment

    • Advantages: High specificity, can detect multiple reaction intermediates

    • Limitations: Requires specialized equipment, challenging quantification

For optimal results, researchers should consider co-expression and co-purification of both gatD and gatE subunits, as the complete GatDE complex exhibits higher activity than individual subunits . Additionally, it's essential to include control reactions lacking either ATP or glutamine to confirm the specificity of the observed activity.

Assay TypeSensitivityThroughputEquipment RequirementsAdvantages
SpectrophotometricHighMediumSpectrophotometerReal-time kinetics
TLC-RadioactiveVery HighLowRadiation facilityDirect measurement
Mass SpectrometryHighLow-MediumMS instrumentPrecise identification
HPLC-basedMedium-HighLow-MediumHPLC systemGood quantification

How do structural features of gatE influence its interaction with gatD and what mutations might reveal about functional domains?

The gatE subunit in Methanococcus maripaludis functions as part of the GatDE complex, which allows for the correct formation of Gln-tRNA(Gln) through transamidation of misacylated Glu-tRNA(Gln) . Understanding the structural determinants of gatE-gatD interactions provides insights into archaeal-specific translation mechanisms:

  • Key interaction domains:

    • The N-terminal domain of gatE likely contains residues critical for gatD recognition

    • The central catalytic domain houses residues involved in ATP binding and activation of the phospho-Glu-tRNA(Gln) intermediate

    • The C-terminal domain potentially contributes to tRNA binding specificity

  • Mutation strategies to probe functional domains:

    • Alanine-scanning mutagenesis of conserved residues can identify essential catalytic and binding sites

    • Domain swapping with homologous proteins can identify regions conferring specificity for glutamate vs. aspartate substrates

    • Site-directed mutagenesis targeting predicted ATP-binding motifs can elucidate the energetics of the reaction

  • Interaction analysis methods:

    • Size-exclusion chromatography to assess complex formation

    • Surface plasmon resonance to measure binding kinetics

    • Isothermal titration calorimetry to determine thermodynamic parameters of gatE-gatD interactions

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

Research has shown that the GatDE system in M. maripaludis is specific for glutamate and does not act on aspartate , which distinguishes it from some bacterial systems. Mutations targeting this substrate specificity could provide valuable insights into the evolutionary divergence of tRNA charging mechanisms across different domains of life.

What evolutionary insights can comparative genomic analysis of the gatE gene provide about archaeal translation systems?

Comparative genomic analysis of the gatE gene from Methanococcus maripaludis offers valuable insights into the evolution of archaeal translation systems and broader questions about the diversity of aminoacyl-tRNA formation pathways. Key research directions include:

  • Phylogenetic distribution patterns:

    • The gatE gene represents an archaeal-specific innovation for tRNA charging

    • Analysis of gatE distribution across the Euryarchaeota (including methanogens and extreme halophiles) reveals conservation patterns that reflect essential functions

    • Comparison with bacterial systems (which typically use GatCAB) highlights domain-specific solutions to the same biochemical challenge

  • Sequence conservation analysis:

    • Highly conserved motifs likely represent functional domains essential for catalysis or tRNA recognition

    • Variable regions may indicate lineage-specific adaptations to different environmental conditions

    • Comparative analysis with the related gatD subunit can identify co-evolving residues that maintain functional interactions

  • Horizontal gene transfer considerations:

    • While M. maripaludis shows evidence of lateral gene transfer from distant lineages, the core translation machinery including gatE likely maintains vertical inheritance patterns

    • Analysis of codon usage and G+C content (which averages 33.1% in M. maripaludis) can help identify genes with potential lateral transfer history

  • Evolutionary model implications:

    • The presence of the GatDE system specifically in Archaea supports models of early divergence in translation systems

    • The absence of glutaminyl-tRNA synthetase in organisms with gatE suggests an ancient solution to the challenge of accurate glutamine incorporation in proteins

This research direction contributes to our understanding of the evolutionary history of translation systems and provides insight into the diversity of solutions that have evolved for accurate protein synthesis across domains of life.

How can genetic manipulation techniques be optimized for studying gatE function in native M. maripaludis?

Studying gatE function within its native context in Methanococcus maripaludis presents unique challenges due to the specialized growth requirements of methanogens and the essential nature of translation machinery components. Advanced genetic manipulation approaches include:

  • Conditional expression systems:

    • Development of tightly controlled inducible promoters for M. maripaludis

    • Implementation of degron-based protein depletion systems to study essential genes

    • Creation of temperature-sensitive variants to allow functional studies under permissive and non-permissive conditions

  • Homologous recombination strategies:

    • Precise genome editing using techniques adapted from those established for glnA in M. maripaludis

    • Creation of marker-less mutations using counterselectable markers

    • Implementation of CRISPR-Cas9 systems adapted for methanogens

  • Gene transfer considerations:

    • Utilize transduction-like gene transfer methods similar to those observed in the related methanogen M. voltae, where small DNA fragments (~4.4-kbp) protected from DNase can transform cells

    • Optimize electroporation protocols specifically for M. maripaludis considering its unique cell wall architecture

    • Develop shuttle vectors with appropriate replication origins for stable maintenance

  • Expression monitoring approaches:

    • Implementation of reporter gene systems functional in the archaeal cellular context

    • Development of protocols for RNA-seq and ribosome profiling in methanogens

    • Establishment of methods for detecting protein-tRNA interactions in vivo

Given that attempts to create null mutants for essential genes like glnA in M. maripaludis have failed , researchers should anticipate that gatE may similarly be essential. Therefore, approaches that allow modulation rather than complete elimination of function will likely be most successful. The nitrogen regulation systems identified in M. maripaludis, including specific operator sequences that mediate repression , could potentially be adapted to create controllable expression systems for studying gatE function.

What are common challenges in obtaining active recombinant gatE and how can they be addressed?

Researchers frequently encounter several challenges when working with recombinant gatE from Methanococcus maripaludis. These challenges and their solutions include:

  • Protein solubility issues:

    • Challenge: Formation of inclusion bodies due to improper folding

    • Solutions:

      • Lower expression temperature (16-18°C) post-induction

      • Co-expression with archaeal-specific chaperones

      • Use fusion partners (SUMO, MBP, thioredoxin) to enhance solubility

      • Implement DoE approaches to systematically optimize expression conditions

  • Loss of activity during purification:

    • Challenge: Recombinant gatE may lose activity during isolation steps

    • Solutions:

      • Include stabilizing agents like glycerol (5-10%) and reducing agents

      • Minimize purification steps and processing time

      • Consider co-expression with partner protein gatD for stability

      • Test multiple buffer compositions using fractional factorial design

  • Lack of functional interaction with gatD:

    • Challenge: Recombinant gatE may not form proper complex with gatD

    • Solutions:

      • Co-expression of both subunits to allow complex formation during synthesis

      • In vitro reconstitution under controlled conditions

      • Introduction of linkers or tags that do not interfere with interaction surfaces

  • Codon usage bias:

    • Challenge: Low expression due to M. maripaludis' low G+C content (33.1%)

    • Solutions:

      • Codon optimization for expression host

      • Use of strains with supplemental tRNAs for rare codons

      • Sequential increase of rare codons from N- to C-terminus to facilitate translation

ChallengePrimary IndicatorFirst-line SolutionAdvanced Solution
Inclusion bodiesProtein in pellet fractionLower induction temperatureDoE optimization of expression
Loss of activityDecreased enzyme functionAdd stabilizing agentsMinimize purification steps
Poor gatD interactionLow complex formationCo-expression strategyInterface engineering
Codon biasLow expression levelUse Rosetta or similar strainsCodon optimization

Systematic application of Design of Experiments approaches allows researchers to efficiently identify optimal conditions by testing multiple variables simultaneously rather than the less efficient one-factor-at-a-time method . This is particularly valuable for complex archaeal proteins like gatE.

How can researchers validate that recombinant gatE retains its native substrate specificity?

Ensuring that recombinant gatE from Methanococcus maripaludis maintains its natural substrate specificity for glutamate (and not aspartate) is critical for both basic research and potential biotechnological applications. Validation approaches include:

  • Comparative activity assays:

    • Test activity with both Glu-tRNA(Gln) and Asp-tRNA(Asn) substrates

    • Measure reaction rates under identical conditions to quantify specificity

    • Expected result: Significant activity only with Glu-tRNA(Gln) substrates

  • Competitive inhibition analysis:

    • Perform reactions with labeled Glu-tRNA(Gln) in the presence of increasing concentrations of Asp-tRNA(Asn)

    • Quantify inhibition constants to determine binding preferences

    • Plot Lineweaver-Burk transformations to identify inhibition patterns

  • Structural biology approaches:

    • Use hydrogen-deuterium exchange mass spectrometry to map substrate binding regions

    • Employ molecular docking simulations to predict binding of different substrates

    • If possible, obtain crystal structures with bound substrates or substrate analogs

  • Mutation analysis:

    • Identify and mutate residues predicted to be involved in substrate discrimination

    • Measure changes in specificity ratios (Glu vs. Asp preference)

    • Use the results to map the substrate specificity-determining regions

A comprehensive validation protocol might include:

Validation LevelMethodExpected Outcome for Native-like Specificity
BasicActivity with Glu vs. Asp substrates>10-fold higher activity with Glu substrate
IntermediateKinetic parameter determinationLower Km and higher kcat for Glu substrate
AdvancedCompetition assaysNo significant inhibition by Asp substrate
DefinitiveStructural analysisSpecific interactions with Glu substrate

What data analysis approaches are most appropriate for interpreting gatE functional studies?

Interpreting data from functional studies of recombinant M. maripaludis gatE requires sophisticated analysis approaches that account for the complex nature of enzymatic reactions in the GatDE system. Recommended analytical methods include:

  • Enzyme kinetics analysis:

    • Apply Michaelis-Menten modeling to determine Km, Vmax, and kcat parameters

    • Use integrated rate equations for analyzing time-course data

    • Implement global fitting approaches for complex reaction mechanisms

    • Consider cooperative binding models if appropriate

  • Statistical validation approaches:

    • For DoE-based optimization studies, use response surface methodology to identify optimal conditions

    • Apply ANOVA to determine significance of experimental factors

    • Use outlier detection methods appropriate for biochemical data

    • Calculate confidence intervals for derived kinetic parameters

  • Visual data representation:

    • Create substrate saturation curves with appropriate error representation

    • For multiple-substrate reactions, use 3D plots to visualize interdependencies

    • Implement heat maps for visualizing extensive condition screening results

    • Use radar plots for comparing multiple performance metrics across variants

  • Advanced comparative analysis:

    • Apply principal component analysis to identify key variables in complex datasets

    • Use hierarchical clustering to identify patterns in variant performance

    • Implement machine learning approaches to predict effects of mutations

    • Develop structure-function relationship models when structural data is available

What are the most promising applications of recombinant gatE in synthetic biology and biotechnology?

Recombinant gatE from Methanococcus maripaludis offers several intriguing applications in synthetic biology and biotechnology, leveraging its unique role in the archaeal-specific GatDE transamidation system . Promising research directions include:

  • Expanded genetic code applications:

    • Engineering gatE to accommodate non-canonical amino acids could enable site-specific incorporation of novel functionalities

    • Development of orthogonal translation systems using modified gatE for synthetic cellular compartmentalization

    • Creation of synthetic cells with archaeal-type translation machinery to explore alternative evolutionary paths

  • Protein engineering platforms:

    • Utilizing the substrate specificity of gatE to develop new approaches for post-translational modification

    • Engineering gatE variants with altered specificity to enable novel amino acid incorporations

    • Development of cell-free protein synthesis systems with archaeal components for difficult-to-express proteins

  • Fundamental research tools:

    • Using reconstituted GatDE systems as tools to study the evolution of the genetic code

    • Development of gatE-based biosensors for detecting misacylated tRNAs in vivo

    • Creating minimal translation systems incorporating archaeal components to understand the core requirements for protein synthesis

  • Therapeutic and diagnostic applications:

    • Exploring gatE inhibitors as potential narrow-spectrum antimicrobials targeting archaeal pathogens

    • Development of diagnostic tools based on unique properties of archaeal translation components

    • Engineering delivery systems for therapeutic proteins using archaeal protein production machinery

This research direction benefits from the distinctive properties of M. maripaludis as a genetically tractable model organism and from the unique characteristics of its GatDE system, particularly its specificity for glutamate over aspartate substrates . Systematic application of Design of Experiments approaches can accelerate progress by optimizing complex experimental parameters more efficiently than traditional methods .

How might structural modifications of gatE be engineered to create variants with novel substrate specificities?

Engineering gatE from Methanococcus maripaludis to accept novel substrates represents an exciting frontier in protein engineering. Strategic approaches to create gatE variants with modified specificities include:

  • Rational design based on structural insights:

    • Target residues in the predicted substrate binding pocket using comparative structural models

    • Implement conservative substitutions to gradually shift specificity

    • Create focused libraries targeting 3-5 residues simultaneously in critical regions

    • Use computational prediction to identify mutations that might accommodate alternative substrates

  • Directed evolution strategies:

    • Develop high-throughput screening systems to detect novel activities

    • Implement PACE (Phage-Assisted Continuous Evolution) for continuous selection of improved variants

    • Apply neutral drift approaches to increase evolutionary plasticity

    • Use computational design to guide library construction

  • Domain swapping approaches:

    • Exchange domains between gatE and related enzymes with different specificities

    • Create chimeric proteins combining elements from archaeal and bacterial transamidation systems

    • Graft specificity-determining loops from related enzymes

    • Engineer fusion proteins combining different catalytic activities

  • Active site remodeling:

    • Enlarge binding pockets to accommodate bulkier substrates

    • Modify electrostatic environment to alter charged substrate preferences

    • Introduce new catalytic residues to enable novel reaction chemistries

    • Stabilize transition states for non-native reactions through hydrogen bonding networks

Such engineering efforts benefit from the understanding that the native GatDE system in M. maripaludis is highly specific for glutamate and does not act on aspartate , providing a clear baseline for measuring changes in specificity. A systematic DoE approach can efficiently optimize experimental conditions across multiple variables simultaneously, reducing the time and resources required compared to traditional optimization methods .

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.