Recombinant Treponema denticola Leucine--tRNA ligase (leuS), partial

Shipped with Ice Packs
In Stock

Description

Overview

Recombinant Treponema denticola Leucine--tRNA ligase (LeuS), partial, is a synthesized form of the Leucine--tRNA ligase enzyme found in the bacterium Treponema denticola . T. denticola is a spirochetal bacterium associated with periodontal diseases . Leucine--tRNA ligase, also known as leucyl-tRNA synthetase (LRS), is an enzyme that catalyzes the attachment of leucine to its corresponding tRNA molecule, a crucial step in protein synthesis .

Leucyl-tRNA Synthetase Function

Leucyl-tRNA synthetase (LRS) is an essential enzyme that ensures the accurate translation of the genetic code . Its primary function involves catalyzing the covalent attachment of leucine to its cognate tRNALeu^{\text{Leu}} . This process is vital for incorporating leucine into polypeptide chains during protein synthesis.

Beyond its role in translation, LRS can also hydrolyze mischarged tRNAs through an editing mechanism . Furthermore, in humans, cytosolic leucyl-tRNA synthetase (hcLRS) functions as a leucine sensor in the rapamycin complex 1 (mTORC1) pathway .

Treponema denticola and LrrA

Treponema denticola is a bacterium that contains a leucine-rich repeat protein, LrrA . LrrA contains eight consensus tandem repeats of 23 amino acid residues . LrrA is associated with the extracytoplasmic fraction of T. denticola and expresses multifunctional properties . LrrA plays a role in the attachment and penetration of human epithelial cells and coaggregation with Tannerella forsythensis .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. 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% and may serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
leuS; TDE_2339; Leucine--tRNA ligase; EC 6.1.1.4; Leucyl-tRNA synthetase; LeuRS
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Treponema denticola (strain ATCC 35405 / CIP 103919 / DSM 14222)
Target Names
leuS
Uniprot No.

Target Background

Database Links

KEGG: tde:TDE2339

STRING: 243275.TDE2339

Protein Families
Class-I aminoacyl-tRNA synthetase family
Subcellular Location
Cytoplasm.

Q&A

What is the genomic organization of the Leucine--tRNA ligase (leuS) gene in Treponema denticola?

The leuS gene in T. denticola encodes Leucine--tRNA ligase, an essential enzyme for protein synthesis that catalyzes the attachment of leucine to its cognate tRNA. While the search results don't specifically detail the leuS gene organization, T. denticola genomic analysis typically reveals that housekeeping genes like leuS are conserved across strains. For instance, the LrrA gene in T. denticola ATCC 35405 has been fully sequenced and characterized, but was not identified in strain ATCC 33520, demonstrating strain-specific genetic variations that researchers must consider when working with T. denticola genes . When designing primers for leuS amplification, researchers should analyze conserved regions across multiple T. denticola reference genomes to ensure successful gene isolation.

How does the structure and function of T. denticola Leucine--tRNA ligase compare to other bacterial aminoacyl-tRNA synthetases?

T. denticola Leucine--tRNA ligase belongs to the class I aminoacyl-tRNA synthetase family, characterized by a Rossmann fold catalytic domain. While specific structural information for T. denticola leuS is not detailed in the search results, bacterial Leucine--tRNA ligases typically contain an aminoacylation domain and an editing domain to ensure fidelity of translation. Similar to other bacterial tRNA synthetases, the enzyme likely follows a two-step reaction: first activating leucine with ATP to form leucyl-adenylate, then transferring the leucyl group to the appropriate tRNA. The detailed molecular interactions would be comparable to the characterized leucyl-tRNA synthetases from other bacterial species, though with sequence variations that may reflect adaptation to the unique physiological environment of oral spirochetes like T. denticola .

What are the optimal conditions for expressing recombinant T. denticola leuS in E. coli expression systems?

For optimal expression of recombinant T. denticola leuS in E. coli, researchers should consider the following parameters:

  • Expression vector selection: Vectors with strong, inducible promoters (T7, tac) are recommended.

  • E. coli host strain: BL21(DE3) derivatives are often optimal for recombinant protein expression.

  • Induction conditions: 0.1-1.0 mM IPTG at OD600 0.6-0.8, with post-induction growth at 16-25°C for 12-18 hours to minimize inclusion body formation.

  • Growth media: Enriched media such as 2xYT or Terrific Broth supplemented with appropriate antibiotics.

Similar methodologies have been successfully applied for other T. denticola recombinant proteins. For example, when expressing the LrrA protein, researchers constructed expression plasmids by amplifying specific gene regions using PCR with designed primers containing appropriate restriction sites (e.g., SalI and BglII) . Temperature optimization is particularly important, as lower temperatures (16-20°C) often improve solubility of recombinant proteins while maintaining reasonable expression levels.

What purification strategies are most effective for recombinant T. denticola leuS?

Effective purification of recombinant T. denticola leuS typically involves a multi-step approach:

  • Affinity chromatography: His-tagged leuS can be purified using Ni-NTA columns with imidazole gradient elution (50-300 mM).

  • Ion exchange chromatography: Based on the theoretical pI of leuS, either cation or anion exchange can further remove contaminants.

  • Size exclusion chromatography: As a final polishing step to achieve >95% purity and remove aggregates.

  • Buffer optimization: Typical buffer includes 20-50 mM Tris-HCl or phosphate buffer (pH 7.5-8.0), 100-300 mM NaCl, 1-5 mM DTT or 2-ME, and 5-10% glycerol for stability.

For related T. denticola proteins like LrrA, researchers have successfully utilized similar chromatographic approaches with adaptations specific to the protein's biochemical properties . Optimization of salt concentration is particularly important to maintain enzyme activity while preventing aggregation. Consider including protease inhibitors in early purification steps to prevent degradation by E. coli proteases.

How can I verify the identity and activity of purified recombinant T. denticola leuS?

To verify the identity and activity of purified recombinant T. denticola leuS, implement the following analytical techniques:

  • Protein identity verification:

    • SDS-PAGE analysis (expected molecular weight)

    • Western blot with anti-His antibodies or specific anti-leuS antibodies

    • Mass spectrometry (MALDI-TOF or LC-MS/MS) for peptide mass fingerprinting

    • N-terminal sequencing

  • Functional activity assessment:

    • Aminoacylation assay measuring the attachment of [14C]-leucine to tRNA

    • ATP-PPi exchange assay to assess amino acid activation

    • Thermal shift assays to evaluate protein stability

Similar verification methods have been employed for other T. denticola recombinant proteins, such as LrrA, where researchers confirmed protein expression through Western blot analysis . For aminoacylation activity, a standard assay involves incubating the enzyme with ATP, tRNA, and radioactively labeled leucine, then measuring the formation of Leu-tRNALeu by TCA precipitation and scintillation counting.

What experimental approaches are most effective for studying structure-function relationships in T. denticola leuS?

For investigating structure-function relationships in T. denticola leuS, researchers should employ a combination of complementary approaches:

  • Site-directed mutagenesis: Systematically mutate conserved residues in catalytic domains, ATP-binding sites, and tRNA-binding regions to assess their roles in enzyme function.

  • Structural biology techniques:

    • X-ray crystallography of leuS alone and in complex with substrates

    • Cryo-EM for visualizing larger complexes

    • NMR for studying dynamic regions and ligand interactions

  • Enzymatic assays with variants:

    • Kinetic analysis comparing wild-type and mutant forms

    • Thermal stability assessments using differential scanning fluorimetry

    • Binding affinity measurements using isothermal titration calorimetry

  • Computational approaches:

    • Molecular dynamics simulations to study conformational changes

    • Homology modeling using related aminoacyl-tRNA synthetases as templates

Similar approaches have been applied to study T. denticola proteins like TvpA, which underwent structural analysis revealing a C-lectin fold that provided insights into its potential functions despite low sequence similarity to functionally characterized proteins . When designing a structure-function study, carefully plan the experimental design to avoid pitfalls such as non-factorial designs that would complicate data analysis and interpretation .

How does T. denticola leuS contribute to pathogenicity in periodontal disease models?

While the direct role of leuS in T. denticola pathogenicity is not explicitly detailed in the search results, we can draw insights from related research on T. denticola virulence factors:

T. denticola leuS likely contributes to pathogenicity through multiple mechanisms:

  • Essential role in protein synthesis: As a critical housekeeping enzyme, leuS enables translation of virulence factors necessary for host colonization and immune evasion.

  • Potential moonlighting functions: Many aminoacyl-tRNA synthetases have secondary functions beyond translation, possibly including:

    • Involvement in biofilm formation

    • Immune modulation

    • Adhesion to host tissues

  • Contribution to stress adaptation: Proper protein synthesis under the stressful conditions of periodontal pockets (oxidative stress, pH fluctuations) requires fully functional tRNA synthetases.

The importance of protein-protein interactions in T. denticola pathogenicity is demonstrated by LrrA, which plays key roles in attachment to human epithelial cells and coaggregation with Tannerella forsythensis, both critical for virulence . Similarly, T. denticola has been shown to form specific interrelationships with P. gingivalis in the periodontal pocket, enhancing virulence in polymicrobial infections . To directly study leuS contribution to pathogenicity, researchers could develop leuS conditional mutants and evaluate their effects on virulence in cell culture and animal models.

What approaches should be used to develop specific inhibitors targeting T. denticola leuS for potential therapeutic applications?

Developing specific inhibitors targeting T. denticola leuS would require a systematic approach:

  • Target validation:

    • Generate conditional leuS mutants to confirm essentiality

    • Perform complementation studies with orthologs to identify unique features

  • High-throughput screening platforms:

    • Design aminoacylation assays adaptable to 384-well format

    • Develop fluorescence-based activity assays for rapid screening

  • Structure-based drug design:

    • Obtain high-resolution crystal structures of T. denticola leuS

    • Identify unique binding pockets absent in human leucyl-tRNA synthetase

    • Perform in silico docking of compound libraries

  • Lead optimization strategies:

    • Medicinal chemistry to improve selectivity, potency, and physicochemical properties

    • Assessment of effects on other oral microbiome species to ensure specificity

  • Delivery systems for periodontal application:

    • Local delivery formulations (gels, films, fibers)

    • Controlled-release systems for sustained inhibitor presence in periodontal pockets

This approach draws on the understanding of T. denticola's role in periodontal diseases and its interactions with other periodontal pathogens like P. gingivalis and T. forsythensis . When developing such inhibitors, researchers should carefully design experiments that account for potential heteroscedasticity in treatment conditions, which can reduce statistical power if not properly addressed in the experimental design .

How can molecular dynamics simulations enhance our understanding of T. denticola leuS substrate recognition and catalytic mechanism?

Molecular dynamics (MD) simulations provide valuable insights into T. denticola leuS function through several approaches:

  • Binding pocket dynamics:

    • Simulations of apo-enzyme flexibility

    • Analysis of conformational changes upon substrate binding

    • Identification of water-mediated interactions critical for substrate recognition

  • Substrate recognition mechanisms:

    • Free energy calculations to quantify binding energetics

    • Steered MD to map substrate entry/exit pathways

    • Identification of key residues for leucine discrimination

  • Catalytic mechanism elucidation:

    • QM/MM approaches to model the reaction coordinate

    • Calculation of activation barriers for rate-limiting steps

    • Investigation of proton transfer events during catalysis

  • Enzyme specificity determination:

    • Comparative simulations with non-cognate amino acids

    • Analysis of editing domain dynamics during proofreading

    • Prediction of specificity-determining residues

  • Simulation validation protocols:

    • Correlation with experimental mutagenesis data

    • Comparison with kinetic measurements

    • Validation against available structural information

This computational approach is particularly valuable when working with proteins like T. denticola leuS where direct structural information may be limited. Similar computational approaches have been applied to understand the function of other T. denticola proteins, such as TvpA, which was found to share structural similarities with Mtd despite low sequence similarity . When designing MD studies, researchers should carefully plan analyses that allow for statistical comparison between conditions while avoiding the pitfalls of multiple hypothesis testing without appropriate corrections .

What are the challenges and solutions for studying interactions between T. denticola leuS and other proteins in the context of multi-protein complexes?

Studying T. denticola leuS interactions within multi-protein complexes presents several challenges with corresponding solutions:

Challenges:

  • Transient interactions: Many aminoacyl-tRNA synthetase interactions are dynamic and difficult to capture.

  • Complex stability: Multi-protein complexes may dissociate during purification.

  • Artificial interactions: Recombinant tags may introduce non-physiological binding.

  • Membrane association: Potential membrane localization complicates analysis.

  • Host-specific factors: E. coli expression may lack T. denticola-specific interaction partners.

Solutions:

  • Proximity-based in vivo approaches:

    • FRET/BRET for live cell interaction monitoring

    • Proximity labeling (BioID, APEX) to identify neighboring proteins

    • Chemical crosslinking followed by MS (XL-MS) to capture transient interactions

  • Advanced purification strategies:

    • Tandem affinity purification with physiological buffer conditions

    • GraFix method for stabilizing complexes with gradient fixation

    • Native mass spectrometry for intact complex analysis

  • Cryo-electron microscopy:

    • Single-particle analysis for structural characterization

    • Tomography for visualizing complexes in cellular context

  • Functional validation methods:

    • Mutational analysis of predicted interaction interfaces

    • Co-localization studies in T. denticola using fluorescent tags

    • Bacterial two-hybrid assays with T. denticola proteins

This approach builds upon techniques used to study protein-protein interactions in T. denticola, such as those demonstrating that LrrA binds specifically to a portion of the Tannerella forsythensis leucine-rich repeat protein BspA . When designing interaction studies, researchers should consider partial within-subjects designs that account for condition-specific data availability and employ appropriate multilevel statistical models for analysis .

What are the key considerations for designing experiments to study T. denticola leuS function in vitro?

When designing experiments to study T. denticola leuS function in vitro, researchers should consider:

  • Enzyme preparation and stability:

    • Optimize buffer conditions (pH 7.0-8.0, 50-200 mM NaCl, 5-10% glycerol)

    • Include stabilizing agents (DTT/TCEP, Mg2+)

    • Determine protein stability timeline at various temperatures

    • Consider flash-freezing aliquots to prevent repeated freeze-thaw cycles

  • Substrate selection and quality:

    • Use highly purified tRNALeu (either native or in vitro transcribed)

    • Ensure ATP purity and stability

    • Consider isotopically labeled substrates for sensitive detection

  • Assay design principles:

    • Include appropriate positive and negative controls

    • Design concentration ranges to capture Michaelis-Menten kinetics

    • Implement multiple detection methods (radioactive, HPLC, coupled-enzyme)

    • Validate assay linearity, precision, and dynamic range

  • Environmental variables:

    • Test temperature range (25-42°C)

    • Evaluate pH optima and ionic strength effects

    • Consider oxygen sensitivity and reducing environment

Researchers should consult a statistician during experimental design to avoid common pitfalls . Ensuring factorial designs where every combination of experimental factors is tested will facilitate complete interactional model fitting. Pre-planning the analysis approach will help identify potential complications before data collection begins .

How should I design a knockout/complementation system to study leuS function in T. denticola?

Designing an effective knockout/complementation system for T. denticola leuS requires careful consideration of this gene's essential nature:

  • Conditional knockout strategy:

    • As leuS is likely essential, develop an inducible promoter system

    • Consider tetracycline-responsive elements or riboswitch-based systems

    • Create a merodiploid strain with a complementary copy before knockout

  • Complementation strategy:

    • Clone wild-type leuS into a T. denticola shuttle vector (like pKMR4PE)

    • Include native promoter and Shine-Dalgarno sequence

    • Transform into conditional knockout strain

    • Verify expression by Western blot

  • Phenotypic analysis:

    • Growth curve analysis under various conditions

    • Microscopic examination for morphological changes

    • Stress response evaluation

    • Virulence factor expression assessment

This approach draws from successful strategies used for creating gene knockouts in T. denticola, such as the lrrA-inactivated mutant, where researchers amplified a portion of the target gene, cloned it into pCR2.1, inserted an ermF-ermAM cassette, and transformed linearized plasmid into T. denticola . For complementation, similar approaches using the T. denticola shuttle vector system have proved effective . When designing such experiments, ensure full factorial designs to avoid complications in data analysis and interpretation .

What controls and validations are essential when studying the impact of environmental factors on T. denticola leuS expression and activity?

When studying environmental influences on T. denticola leuS expression and activity, implement these essential controls and validations:

Experimental Controls:

  • Biological replicate controls:

    • Independent bacterial cultures (n≥3)

    • Technical replicates within each biological replicate

  • Reference condition controls:

    • Standard growth conditions as baseline

    • Vehicle controls for chemical treatments

    • Mock treatment controls

  • Internal gene/protein controls:

    • Constitutively expressed housekeeping genes (16S rRNA, gyrA)

    • Non-responsive proteins as negative controls

    • Known responsive genes as positive controls

Validation Approaches:

  • Expression analysis validation:

    • Multiple quantification methods (qRT-PCR, RNA-Seq)

    • Protein-level confirmation (Western blot, proteomics)

    • Promoter activity assessment (reporter constructs)

  • Activity measurement validation:

    • Multiple activity assay methods

    • Dose-response relationships

    • Time-course studies to capture dynamics

  • Physiological relevance validation:

    • Comparison with in vivo conditions

    • Co-culture experiments to assess polymicrobial effects

    • Host cell interaction studies

When designing these experiments, researchers should be aware of expected violations like heteroscedasticity between treatment conditions, which affects statistical power. Planning for these issues by recruiting additional participants or samples to compensate for the degrees-of-freedom reduction in heteroscedastic tests is essential . Additionally, ensuring complete factorial designs will prevent challenges in interactional model fitting .

What approaches can be used to study the impact of T. denticola leuS mutations on bacterial fitness and virulence?

To study the impact of T. denticola leuS mutations on fitness and virulence, researchers can implement these approaches:

  • Site-directed mutagenesis strategy:

    • Target conserved catalytic residues

    • Modify specificity-determining residues

    • Create chimeric enzymes with domains from other species

    • Design mutations based on structural predictions

  • Fitness assessment methodologies:

    • Growth curve analysis (doubling time, lag phase, maximum density)

    • Competition assays with wild-type strains (fitness index calculation)

    • Stress survival studies (pH, oxidative, nutrient limitation)

    • Biofilm formation capacity assessment

  • Virulence characteristic evaluation:

    • Motility assays (swarming on semi-solid media)

    • Attachment to epithelial cells (similar to LrrA studies )

    • Coaggregation with other periodontal pathogens

    • Penetration of tissue models

  • In vivo virulence models:

    • Mouse abscess model with defined bacterial challenge doses

    • Polymicrobial infection models with P. gingivalis (assessing synergistic effects )

    • Host immune response characterization

  • Multi-omics approach to system-wide effects:

    • Transcriptome analysis of mutant vs. wild-type

    • Proteome changes in response to mutation

    • Metabolomic shifts indicating compensatory mechanisms

This approach builds on existing knowledge about T. denticola virulence factors, such as LrrA, which when mutated showed reduced attachment to HEp-2 cells, decreased coaggregation with T. forsythensis, and attenuated tissue penetration . When designing these experiments, researchers should consider potential interactions between factors and employ full factorial designs to enable complete statistical modeling .

How can I design experiments to compare the structural and functional conservation of leuS between T. denticola and related oral pathogens?

To compare leuS conservation across oral pathogens, implement this experimental design:

  • Sequence-based comparative analysis:

    • Multiple sequence alignment of leuS from T. denticola, T. pallidum, P. gingivalis, and other oral pathogens

    • Phylogenetic tree construction to establish evolutionary relationships

    • Conservation mapping onto known aminoacyl-tRNA synthetase structures

    • Identification of species-specific insertions/deletions

  • Structural comparison approaches:

    • Homology modeling of each species' leuS

    • Superimposition analysis to identify conserved structural elements

    • Binding pocket comparison focusing on substrate specificity

    • Molecular dynamics simulations to compare conformational flexibility

  • Functional complementation studies:

    • Cross-species genetic complementation assays

    • Expression of heterologous leuS genes in T. denticola conditional mutants

    • Chimeric enzyme construction and activity assessment

    • Domain swapping experiments to identify functional equivalence

  • Biochemical property comparison:

    SpeciesKm (Leu)Km (ATP)Km (tRNA)kcatTemperature OptimumpH Optimum
    T. denticolax μMy μMz μMa s⁻¹b°CpH c
    T. pallidum..................
    P. gingivalis..................
  • Inhibitor sensitivity profiling:

    • Screen candidate inhibitors against leuS from multiple species

    • Determine IC50 values for each organism

    • Establish structure-activity relationships

    • Identify species-selective compounds

This approach draws inspiration from comparative studies of other T. denticola proteins, such as the leucine-rich repeat protein LrrA, which was compared to similar proteins in T. pallidum (TpLRR) and T. forsythensis (BspA) . When designing cross-species comparison experiments, researchers should ensure full factorial designs where every combination of species and experimental conditions is tested, avoiding the pitfall of non-factorial designs that prevent full interactional model fitting .

What are the common challenges in expressing and purifying active recombinant T. denticola leuS, and how can they be addressed?

Researchers frequently encounter these challenges when working with recombinant T. denticola leuS:

Challenge 1: Poor expression levels

  • Potential causes: Codon bias, toxic effects, mRNA secondary structure

  • Solutions:

    • Optimize codons for E. coli expression

    • Use specialized E. coli strains (Rosetta, CodonPlus)

    • Employ tightly regulated expression systems

    • Try fusion partners (SUMO, MBP, TrxA) to enhance solubility

Challenge 2: Inclusion body formation

  • Potential causes: Rapid overexpression, improper folding

  • Solutions:

    • Reduce induction temperature (16-20°C)

    • Decrease IPTG concentration (0.1-0.2 mM)

    • Co-express with chaperones (GroEL/ES, DnaK/J)

    • Consider auto-induction media for slower expression

Challenge 3: Enzyme instability

  • Potential causes: Oxidation of cysteines, proteolysis, aggregation

  • Solutions:

    • Include reducing agents (DTT, TCEP) in all buffers

    • Add protease inhibitors during lysis

    • Optimize buffer conditions (pH, salt, glycerol)

    • Process samples rapidly at 4°C

Challenge 4: Low activity of purified enzyme

  • Potential causes: Improper folding, missing cofactors, inhibitory contaminants

  • Solutions:

    • Include Mg²⁺ and K⁺ in activity buffers

    • Remove imidazole completely after affinity purification

    • Test activity with varied tRNA sources

    • Consider native purification conditions

These troubleshooting approaches are informed by general principles of recombinant protein expression and purification, adapted for the specific challenges of T. denticola proteins. When designing optimization experiments, researchers should follow factorial designs to simultaneously evaluate multiple variables and their interactions, avoiding the pitfalls of one-factor-at-a-time approaches that miss important interaction effects .

How should I analyze kinetic data for T. denticola leuS to account for potential allosteric effects and substrate inhibition?

When analyzing kinetic data for T. denticola leuS with potential allosteric effects and substrate inhibition, implement this comprehensive approach:

  • Initial data visualization and model selection:

    • Plot velocity vs. substrate concentration using non-transformed axes

    • Look for characteristic signs of substrate inhibition (decrease at high [S])

    • Check for sigmoidal curves indicating allosteric behavior

    • Consider using the Akaike Information Criterion (AIC) to select the best model

  • Mathematical models for different kinetic behaviors:

    Kinetic BehaviorMathematical ModelWhen to Apply
    Michaelis-MentenV = Vmax[S]/(Km+[S])Hyperbolic curves with no inhibition or cooperativity
    Substrate InhibitionV = Vmax[S]/(Km+[S]+(([S]²)/Ki))Curves showing decreased velocity at high substrate
    Hill EquationV = Vmax[S]ⁿ/(K'+(S)ⁿ)Sigmoidal curves indicating cooperativity
    Mixed ModelV = Vmax[S]ⁿ/(K'+(S)ⁿ+(([S]²)/Ki))Combined allosteric effects and substrate inhibition
  • Statistical approach for parameter estimation:

    • Use nonlinear regression rather than linearization methods

    • Calculate confidence intervals for each parameter

    • Perform residual analysis to validate model fit

    • Consider weighted regression if heteroscedasticity is present

  • Experimental design for detecting allosteric effects:

    • Test effects of potential allosteric effectors at multiple concentrations

    • Perform substrate titrations at different effector concentrations

    • Analyze data with global fitting approaches

    • Consider isothermal titration calorimetry for direct binding measurements

When analyzing such complex kinetic data, researchers should be aware that heteroscedasticity commonly occurs (larger variance at higher substrate concentrations), which affects statistical inference. Using appropriate weighted regression methods and planning for adequate replication to compensate for the reduced statistical power is essential .

What statistical approaches are most appropriate for analyzing differences in leuS expression or activity across different experimental conditions?

For analyzing leuS expression or activity differences across conditions, these statistical approaches are recommended:

  • Exploratory data analysis:

    • Assess normality using Shapiro-Wilk test and Q-Q plots

    • Evaluate homogeneity of variance with Levene's test

    • Identify potential outliers using boxplots and influence diagnostics

    • Create interaction plots to visualize potential effect modifications

  • Statistical test selection framework:

    Data CharacteristicsRecommended TestAdvantagesLimitations
    Normal, homoscedasticANOVA with post-hoc testsPowerful, widely acceptedSensitive to violations of assumptions
    Non-normal or heteroscedasticWelch's ANOVA, Kruskal-WallisRobust to assumption violationsReduced power, limited to simpler designs
    Multiple factors, complete designFactorial ANOVATests main effects and interactionsRequires balanced design for optimal performance
    Multiple factors, incomplete designMixed-effects modelsHandles missing data, nested factorsMore complex to implement and interpret
    Repeated measuresRM-ANOVA or mixed modelsAccounts for within-subject correlationRequires careful consideration of covariance structure
  • Post-hoc testing approach:

    • Tukey's HSD for all pairwise comparisons with controlled FWER

    • Dunnett's test when comparing treatments to a control

    • Bonferroni or Holm-Bonferroni for selected comparisons

    • Consider false discovery rate control for many comparisons

  • Effect size reporting:

    • Report Cohen's d or Hedges' g for pairwise comparisons

    • Include partial η² for ANOVA factors

    • Provide confidence intervals for all effect estimates

    • Consider Bayesian approaches for more nuanced interpretation

This statistical approach is informed by best practices in experimental design and analysis. Researchers should be aware that heteroscedasticity between treatment conditions (e.g., higher variance in some conditions) reduces statistical power by shrinking the degrees-of-freedom in heteroscedastic tests . Planning for these issues by recruiting additional replicates to compensate is recommended .

How can I troubleshoot and optimize T. denticola leuS aminoacylation assays that show inconsistent results?

When troubleshooting inconsistent T. denticola leuS aminoacylation assays, implement this systematic approach:

  • Reagent quality assessment:

    • Verify enzyme purity by SDS-PAGE (>90% homogeneity)

    • Check tRNA integrity by urea PAGE or Bioanalyzer

    • Ensure ATP freshness and proper storage

    • Validate radioactive leucine specific activity and purity

  • Assay component optimization:

    • Titrate enzyme concentration (0.1-500 nM range)

    • Optimize Mg²⁺ concentration (2-20 mM)

    • Test buffer systems (HEPES vs. Tris vs. phosphate)

    • Evaluate additives (BSA, glycerol, reducing agents)

  • Critical procedural controls:

    • Include enzyme-minus controls

    • Perform time-course studies to ensure linearity

    • Implement positive controls with commercial synthetases

    • Include duplicate or triplicate technical replicates

  • Common pitfalls and solutions:

    ProblemPotential CausesSolutions
    High backgroundInefficient washing, filter retentionTry TCA precipitation alternatives, increase wash stringency
    Poor signalEnzyme inactivation, tRNA degradationAdd stabilizers, check for RNase contamination
    Nonlinear kineticsProduct inhibition, enzyme instabilityUse initial rates only, optimize enzyme concentration
    Variable replicatesPipetting errors, temperature variationUse multichannel pipettes, maintain strict temperature control
  • Advanced troubleshooting:

    • Compare multiple detection methods (filter binding, HPLC, scintillation)

    • Validate the assay with commercially available leucyl-tRNA synthetase

    • Consider continuous assays for real-time monitoring

    • Evaluate buffer component interactions using DOE (Design of Experiments)

When designing optimization experiments, researchers should employ full factorial designs to capture interaction effects between factors rather than changing one variable at a time . Additionally, be aware that heteroscedasticity between optimized and unoptimized conditions is common and should be addressed in statistical analyses by using appropriate tests .

What bioinformatic approaches can help identify functional domains and critical residues in T. denticola leuS for targeted mutagenesis studies?

For identifying functional domains and critical residues in T. denticola leuS, implement these bioinformatic approaches:

  • Sequence-based analysis:

    • Multiple sequence alignment with diverse bacterial leuS sequences

    • Conservation scoring to identify invariant residues

    • Identification of class I aminoacyl-tRNA synthetase motifs (HIGH, KMSKS)

    • Disorder prediction to identify flexible regions

  • Structure-based analysis:

    • Homology modeling using closely related leuS structures as templates

    • Molecular docking of leucine, ATP, and tRNA

    • Electrostatic surface mapping to identify potential interaction sites

    • Normal mode analysis to predict conformational changes

  • Integration with experimental data:

    • Map previously identified mutations affecting function from related enzymes

    • Identify residues under selection pressure through evolutionary analysis

    • Correlate with available biochemical data on substrate specificity

  • Specialized tools for critical residue prediction:

    Analysis ApproachRecommended ToolsOutput Information
    Catalytic site predictionCSA, SCANNET, POOLPutative catalytic residues based on structural features
    Ligand binding site analysis3DLigandSite, COACH, SiteMapPotential binding pockets and interacting residues
    Coevolution analysisEVcouplings, GREMLIN, DCACo-evolving residue pairs indicating functional relationships
    Machine learning integrationDeepLeuSite, ProTSPoMML-based predictions of functionally important sites
  • Prioritization strategy for mutagenesis:

    • Focus on residues unique to T. denticola compared to human ortholog

    • Target interface residues in predicted protein-protein interaction sites

    • Mutate residues in the editing domain to alter fidelity

    • Create alanine scanning libraries of predicted functional loops

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.