Recombinant Enterococcus faecalis Tyrosine--tRNA ligase 1 (tyrS1)

Shipped with Ice Packs
In Stock

Description

Function of Tyrosine--tRNA Ligase

Tyrosine--tRNA ligase, also known as tyrosyl-tRNA synthetase, is responsible for the ATP-dependent activation of tyrosine by forming an enzyme-bound tyrosyl-adenylate intermediate. This intermediate then reacts with tyrosine tRNA to form tyrosyl-tRNA, which is essential for the incorporation of tyrosine into proteins during translation .

Role in Tyrosine Metabolism

In Enterococcus faecalis, tyrosine metabolism is closely linked with the production of biogenic amines such as tyramine. The tyrosine decarboxylase (TDC) pathway, which converts tyrosine into tyramine, is a significant aspect of this metabolism. While tyrosyl-tRNA synthetase itself does not directly participate in tyramine production, it can act as a sensor of the intracellular tyrosine pool, potentially influencing the regulation of tyrosine decarboxylation .

Genetic Organization and Regulation

The gene encoding tyrosyl-tRNA synthetase in Enterococcus faecalis is often located upstream of the tyrosine decarboxylase gene (tdc) and is cotranscribed with it and the downstream tyrosine-tyramine antiporter gene (tyrP) in some strains . This cotranscription suggests a coordinated regulation of these genes, potentially influenced by tyrosine availability and other environmental factors.

Research Findings and Data

While specific data on recombinant Enterococcus faecalis Tyrosine--tRNA Ligase 1 (tyrS1) is not readily available, studies on related enzymes and pathways provide insights into its potential functions and regulatory mechanisms. For instance, the expression of the tyrDC gene, which is involved in tyramine production, varies significantly among different strains of Enterococcus faecalis and is influenced by factors such as growth phase and medium composition .

Table: Comparison of Tyramine Production in Different Enterococcal Strains

StrainTyramine Production (μg/mL)Growth Phase
E. faecalis EF37HighExponential
E. faecium FC12HighExponential
E. faecalis ATCC 29212LowerStationary
E. faecium FC12HighStationary

Note: The table illustrates the variability in tyramine production among different enterococcal strains, which may indirectly reflect differences in tyrosine metabolism and regulation.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on purchasing method and location. Consult 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. 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% and can serve as a guideline.
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 for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. Please specify your desired tag type for preferential development.
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-418
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Target Names
tyrS1
Target Protein Sequence
MNIIDELAWRDAINQQTNEEGLRELTENTSISLYCGVDPTGDSMHIGHLIPFMMMKRFQL AGHHPYILIGGGTGTIGDPSGRTTERVLQTMEAVQHNVDSLSNQMKKLFGKDAEVTMVNN YDWLSELSLLDFLRDYGKNFNVNTMLAKDIVASRLESGISFTEFTYQILQSIDFYTLHKK HNIQLQIGGADQWGNITAGLDLIRKKEGPEAKVFGLTIPLMLKADGTKFGKTAGGAIWLD PKKTSPFEFYQFWLNQDDRDVIKYLKFFTFLDKEEIDALAEKVEKEPGKREAQRRLAEEV TRFVHDDAALEEAQKISEALFSGNIKDLTIEEIEQGLEHVPTVEITKDAKNIVDWLVDTE IEPSKRQAREDVSGGAISINGDRVTDLDFAVDPTQHFDGKFVVVRKGKKNYFLAKVMD
Uniprot No.

Q&A

What is Enterococcus faecalis Tyrosine--tRNA ligase 1 (tyrS1)?

Enterococcus faecalis Tyrosine--tRNA ligase 1 (tyrS1) is an aminoacyl-tRNA synthetase-like gene involved in attaching tyrosine to its cognate tRNA molecule. As part of the tyrosine decarboxylase (tdc) gene cluster, tyrS1 plays a role in protein synthesis and potentially in tyrosine metabolism. Unlike conventional aminoacyl-tRNA synthetases, tyrS in E. faecalis exhibits distinct regulatory patterns, being repressed by tyrosine while other genes in the tdc cluster are induced by tyrosine . This enzyme catalyzes a two-step reaction: first activating tyrosine with ATP to form tyrosyl-adenylate, then transferring the tyrosyl group to tRNA^Tyr. The differential regulation of tyrS1 compared to other tdc cluster genes suggests it may have evolved specialized functions related to coordinating protein synthesis with tyrosine metabolism.

How does tyrS1 differ from tyrosyl-tRNA synthetases in other organisms?

Tyrosyl-tRNA synthetases across species share core catalytic functions but exhibit important structural and regulatory differences. E. faecalis tyrS1 is notably part of the tdc cluster involved in tyramine production, unlike most organisms where tyrosyl-tRNA synthetases function independently of decarboxylation pathways . One significant difference is the regulatory pattern: E. faecalis tyrS is repressed by tyrosine, contrasting with the typical constitutive expression of tyrosyl-tRNA synthetases in other bacteria . The structural organization likely parallels that of other bacterial tyrosyl-tRNA synthetases, containing a Rossmann fold for ATP binding, but with specific adaptations for interaction with other tdc cluster components. Research on related TyrRS from Bacillus stearothermophilus has identified a dense cluster of eight residues at the crossing of two alpha-helices that affects enzyme stability and activity, providing insights into potential structural features of E. faecalis tyrS1 . These organism-specific adaptations likely reflect the ecological niche of E. faecalis, allowing coordination between protein synthesis and specialized metabolic pathways.

What experimental methods are used to determine tyrS1 expression and regulation?

Researchers employ multiple complementary approaches to investigate tyrS1 expression and regulation. RT-qPCR analysis has been used to quantify transcriptional changes in tyrS when exposed to varying tyrosine concentrations, revealing that tyrS is repressed by tyrosine unlike other genes in the tdc cluster which are induced . Global transcriptome analysis through microarray experiments has confirmed this differential regulation pattern and identified potential co-regulation with other metabolic pathways . For visualizing expression patterns in vivo, fluorescence analysis using reporter proteins like GFP fused to the tyrS promoter provides spatial and temporal insights into regulation . When investigating regulatory interactions, electrophoretic mobility shift assays (EMSAs) can identify proteins that bind to the tyrS promoter region. Researchers should design experiments that account for multiple variables affecting tyrS1 expression, including growth phase, environmental factors, and cross-talk between different metabolic pathways . Combining these methodologies provides comprehensive understanding of the complex regulation of tyrS1 in E. faecalis.

How is tyrS1 regulated in response to tyrosine levels?

E. faecalis tyrS1 exhibits a unique regulatory response to tyrosine that differs from other genes in the tdc cluster. While the catabolic genes like tdcA, tdcP, and nhac-2 (encoding tyrosine decarboxylase, tyrosine-tyramine antiporter, and Na+/H+ antiporter, respectively) are over-expressed in the presence of tyrosine, tyrS is repressed under the same conditions . This differential regulation has been confirmed through both RT-qPCR and global transcriptome analysis . This pattern suggests a sophisticated regulatory mechanism that coordinates protein synthesis with tyramine production. When tyrosine levels are high, the repression of tyrS1 may limit tyrosine incorporation into proteins while simultaneously inducing the tdc pathway to convert excess tyrosine to tyramine. The molecular mechanisms underlying this regulation might involve transcriptional repressors that bind to the tyrS1 promoter in a tyrosine-dependent manner or RNA structural elements that affect translation efficiency. This regulatory pattern has been demonstrated in multiple E. faecalis strains, suggesting it is a conserved feature of this species . Understanding this regulation is crucial for interpreting the broader metabolic adaptations of E. faecalis to environments rich in tyrosine.

What is the relationship between tyrS1 and biogenic amine production?

Research has revealed a fascinating relationship between tyrS1 and biogenic amine production in E. faecalis. Both tyrS1 and the genes responsible for tyramine production (through the TDC pathway) and putrescine production (through the AGDI pathway) are subject to complex co-regulation . Transcriptome studies have demonstrated that tyrosine not only regulates the tdc cluster (repressing tyrS while inducing catabolic genes) but also induces the transcription of putrescine biosynthesis genes . This cross-pathway regulation suggests a coordinated response to amino acid availability. The modulation of the AGDI route by tyrosine was not observed in a Δtdc mutant strain, indicating that the tdc cluster (which includes tyrS1) is essential for this regulatory interaction . Fluorescence analyses using gfp as a reporter protein confirmed that the AGDI pathway promoter (PaguB) is induced by tyrosine in wild-type but not in tdc mutant strains . This evidence suggests that tyrS1, as part of the tdc cluster, participates in a broader regulatory network coordinating multiple biogenic amine production pathways in response to environmental signals. This coordination may provide metabolic advantages to E. faecalis in specific ecological niches.

What methods can be used to study tyrS1 promoter activity?

Several complementary approaches can be employed to investigate tyrS1 promoter activity and regulation. Fluorescence-based reporter systems, as demonstrated in research on the AGDI pathway, provide valuable tools for visualizing promoter activity in vivo . By fusing the tyrS1 promoter region to reporter genes like gfp, researchers can monitor expression patterns under different conditions and in various genetic backgrounds. This approach was successfully used to demonstrate tyrosine induction of the aguB promoter in wild-type but not in tdc mutant strains . Quantitative assessment of promoter activity can be achieved through luminescence-based reporters (like luxAB or nanoluciferase) that offer high sensitivity and a wide dynamic range. For dissecting the molecular interactions at the promoter, electrophoretic mobility shift assays (EMSAs) can identify proteins that bind to the tyrS1 promoter region under different conditions. DNase footprinting provides complementary information about the specific nucleotides protected by regulatory proteins. Mutagenesis of potential regulatory elements within the promoter, followed by reporter assays, helps identify essential sequences for regulation. In vitro transcription systems using purified components can isolate direct effects on transcription initiation from indirect cellular responses. Together, these methods provide a comprehensive toolkit for unraveling the complex regulation of tyrS1 expression.

What is the optimal protocol for purifying recombinant E. faecalis tyrS1?

Efficient purification of recombinant E. faecalis tyrS1 requires a carefully optimized multi-step protocol. Begin with construct design, incorporating an N-terminal His6-tag for affinity purification and a TEV protease cleavage site for tag removal if necessary. Express the protein in E. coli BL21(DE3) using auto-induction media or IPTG induction (0.5 mM) at lower temperatures (18-20°C overnight) to enhance soluble protein yield. After harvesting cells, resuspend in lysis buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, 5 mM β-mercaptoethanol, 1 mM PMSF) and disrupt by sonication or high-pressure homogenization. Clear lysate by centrifugation (30,000 × g, 45 min, 4°C) before applying to Ni-NTA resin pre-equilibrated with lysis buffer. After washing with buffer containing 20 mM imidazole, elute tyrS1 using an imidazole gradient (50-300 mM). For higher purity, implement ion-exchange chromatography using a Q-Sepharose column with a 0-500 mM NaCl gradient in 50 mM Tris-HCl pH 8.0. As a final polishing step, perform size-exclusion chromatography using a Superdex 200 column equilibrated with storage buffer (25 mM Tris-HCl pH 7.5, 150 mM NaCl, 5% glycerol, 1 mM DTT). Assess protein purity by SDS-PAGE and verify identity by Western blot or mass spectrometry. Activity assays using pyrophosphate exchange or aminoacylation reactions confirm functional integrity. Store purified tyrS1 in small aliquots at -80°C to avoid repeated freeze-thaw cycles.

How can the enzymatic activity of tyrS1 be measured?

Multiple complementary assays can be employed to measure tyrS1 enzymatic activity, each probing different aspects of the aminoacylation reaction. The pyrophosphate exchange assay, successfully used with TyrRS from B. stearothermophilus, measures the first step of the reaction (amino acid activation) . This assay monitors the ATP-dependent exchange of radioactive pyrophosphate into ATP, which occurs when tyrosyl-adenylate formation is reversible. The reaction typically contains 50 mM HEPES pH 7.5, 10 mM MgCl2, 1 mM ATP, 1 mM tyrosine, 0.1 mM [32P]PPi, and purified tyrS1. For measuring complete aminoacylation, the tRNA charging assay tracks the formation of tyrosyl-tRNA^Tyr using either radioisotope-labeled tyrosine or acid precipitation methods. The reaction mixture includes 50 mM HEPES pH 7.5, 10 mM MgCl2, 5 mM ATP, 1 mM tyrosine (or [14C]tyrosine), purified tRNA^Tyr, and tyrS1 enzyme. For continuous monitoring of activity, coupled enzymatic assays link pyrophosphate release to NADH oxidation through auxiliary enzymes. Additionally, thermal shift assays (Thermofluor) can indirectly assess substrate binding by monitoring changes in protein thermal stability upon ligand interaction. For all assays, careful optimization of reaction conditions is essential, including buffer composition, pH (typically 7.5-8.0), divalent cation concentration, and temperature (30-37°C for E. faecalis enzymes). Proper controls including enzyme-free reactions and known inhibitors help validate assay specificity.

What mutagenesis approaches are most effective for studying tyrS1 function?

Site-directed mutagenesis provides powerful tools for dissecting tyrS1 structure-function relationships. Based on studies with related TyrRS enzymes, targeted approaches should focus on key functional regions. For catalytic site analysis, alanine-scanning mutagenesis of conserved motifs (HIGH and KMSKS) can identify essential residues for ATP binding and catalysis. Directed evolution approaches generating libraries of mutations followed by activity screening can identify novel functional variants. For investigating stability determinants, mutations analogous to those studied in B. stearothermophilus TyrRS (such as I52L, M55L, L105V, and T51P) should be created and characterized . Mutations at the dimer interface can probe the importance of quaternary structure for function. Technical implementation should follow a standardized protocol: design mutagenic primers with 15-20 nucleotides flanking the mutation site, perform PCR with high-fidelity polymerase, digest parental DNA with DpnI, transform into competent E. coli, and confirm mutations by sequencing. For complex mutational patterns, gene synthesis offers a more efficient approach. Characterization of mutants should include activity assays (pyrophosphate exchange, aminoacylation), stability assessments (thermal denaturation, chemical denaturation), and structural analysis where possible. Compensatory mutation analysis, as demonstrated with I52L partially compensating for L105V in B. stearothermophilus TyrRS, can reveal intramolecular networks of functional importance .

How can tyrS1 be used to investigate E. faecalis pathogenicity?

Investigating tyrS1 in the context of E. faecalis pathogenicity offers valuable insights into bacterial adaptation and virulence mechanisms. Comparative analysis of tyrS1 sequences and expression patterns between pathogenic clinical isolates (like E. faecalis UW3114) and non-pathogenic strains (such as probiotic E. faecalis Symbioflor 1 or food isolate E. faecalis D27) can reveal strain-specific adaptations . Research has demonstrated that pathogenic strains express unique virulence factors and display distinctive proteomic profiles compared to non-pathogenic counterparts . To investigate tyrS1's potential role in pathogenicity, researchers should: (1) Create tyrS1 knockout mutants and assess impacts on virulence in infection models; (2) Perform RNA-seq analysis comparing gene expression profiles between wild-type and tyrS1 mutants during host interaction; (3) Investigate whether tyrS1-dependent tyrosine metabolism affects the production of virulence factors; (4) Examine tyrS1's potential involvement in stress responses critical for host colonization; and (5) Assess whether tyrS1 participates in horizontal gene transfer networks that contribute to the spread of virulence factors . The fact that E. faecalis has emerged as an important nosocomial pathogen with high levels of antibiotic resistance highlights the importance of understanding all potential contributors to pathogenicity, including specialized metabolic enzymes like tyrS1 .

What is the relationship between tyrS1 and the biogenic amine production pathways?

E. faecalis tyrS1 participates in a sophisticated cross-regulation network connecting tyrosine metabolism with multiple biogenic amine production pathways. Transcriptome studies have revealed that tyrosine not only regulates the tdc cluster (which includes tyrS1) but also induces the agmatine deiminase (AGDI) pathway responsible for putrescine production . This regulatory linkage creates a positive correlation between putrescine biosynthesis and tyrosine concentration . The mechanism of this cross-pathway regulation is particularly intriguing as it depends on the presence of a functional tdc cluster. When the tdc cluster was deleted, tyrosine could no longer induce the AGDI pathway, suggesting that components of the tdc pathway (potentially including tyrS1) mediate this regulatory interaction . Fluorescence analyses using gfp as a reporter confirmed that the promoter of AGDI catabolic genes (PaguB) is induced by tyrosine in wild-type but not in tdc mutant strains . The transcriptional regulator AguR appears to be implicated in this interaction between the two clusters . This complex regulatory network likely allows E. faecalis to coordinate multiple amino acid decarboxylation pathways in response to environmental conditions, potentially contributing to acid tolerance, energy generation through decarboxylation, or other adaptive advantages in specific ecological niches.

Can structural information from related TyrRS enzymes guide inhibitor design for E. faecalis tyrS1?

Structural information from related tyrosyl-tRNA synthetases provides a valuable foundation for structure-based inhibitor design targeting E. faecalis tyrS1. Studies on B. stearothermophilus TyrRS have identified a dense cluster of eight residues at the crossing of two alpha-helices, with four residues not conserved in E. coli TyrRS . This structural feature likely exists in E. faecalis tyrS1 and could represent a species-specific target for selective inhibition. The impact of specific mutations on enzyme stability and activity, as demonstrated with mutations I52L, M55L, L105V, and T51P in B. stearothermophilus TyrRS, provides insights into critical regions for enzyme function . Rational inhibitor design should focus on several key areas: (1) The ATP binding pocket containing the HIGH and KMSKS motifs; (2) The tyrosine binding site that determines amino acid specificity; (3) Interface regions involved in dimerization that could be disrupted by small molecules; and (4) Species-specific structural elements not present in human tyrosyl-tRNA synthetase. Computational approaches including homology modeling, molecular docking, and molecular dynamics simulations can predict binding modes and optimize lead compounds. Fragment-based drug discovery methods could identify initial binding scaffolds that can be elaborated into more potent inhibitors. Given E. faecalis's clinical significance and increasing antibiotic resistance, developing selective tyrS1 inhibitors could provide valuable new therapeutic options.

What bioinformatic approaches help analyze tyrS1 sequence and structure?

Comprehensive bioinformatic analysis of tyrS1 requires multiple complementary approaches. Sequence analysis should begin with multiple sequence alignment of tyrS1 homologs from diverse bacterial species, particularly comparing pathogenic and non-pathogenic strains. BLASTP and HHpred searches identify distant homologs and conserved domains. Phylogenetic analysis using maximum likelihood methods reveals evolutionary relationships and potential horizontal gene transfer events. For structural prediction, homology modeling based on crystal structures of related TyrRS enzymes (like B. stearothermophilus TyrRS) generates initial 3D models . These models can be refined using molecular dynamics simulations to assess stability and conformational flexibility. Structure validation tools like PROCHECK and MolProbity evaluate model quality. Protein-protein interaction prediction identifies potential binding partners within the tdc cluster. For functional analysis, conserved motifs (HIGH, KMSKS) should be mapped onto the structural model, and substrate binding sites predicted using CASTp or similar tools. Conservation analysis using ConSurf highlights functionally important residues. Coevolution analysis methods like direct coupling analysis (DCA) can identify residues that evolve together, suggesting functional coupling. Integration of sequence, structural, and functional predictions provides a comprehensive understanding of tyrS1 biology and guides experimental design for further characterization of this important enzyme.

What methods are most effective for studying tyrS1 interactions with tRNA substrates?

Investigating tyrS1 interactions with tRNA substrates requires specialized techniques that can capture the dynamics and specificity of these interactions. Electrophoretic mobility shift assays (EMSAs) provide a fundamental approach to detecting tyrS1-tRNA binding, with varying concentrations of purified tyrS1 incubated with labeled tRNA^Tyr to determine dissociation constants (Kd). For more detailed binding kinetics, surface plasmon resonance (SPR) or bio-layer interferometry (BLI) enable real-time monitoring of association and dissociation rates. To identify specific nucleotides critical for recognition, RNA footprinting using ribonucleases or chemical probes can map protected regions within the tRNA structure when bound to tyrS1. For structural insights, X-ray crystallography of tyrS1-tRNA complexes provides atomic-resolution information, while cryo-electron microscopy offers visualization of more flexible complexes. To study the aminoacylation reaction specifically, filter binding assays using radiolabeled tyrosine measure the transfer of tyrosine to tRNA over time. Hydroxyl radical probing can map the three-dimensional architecture of the complex in solution. For a comprehensive understanding, these biophysical approaches should be complemented with functional studies using tRNA variants with specific mutations in identity elements. Competitive aminoacylation assays with variant tRNAs reveal the kinetic consequences of specific structural features. Together, these methods provide a multi-faceted view of the critical tyrS1-tRNA interactions that ensure translational fidelity.

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.