Recombinant Bacteroides thetaiotaomicron Shikimate kinase (aroK)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we will prioritize shipping the format currently in stock, please specify any format requirements in your order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. 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 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 forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
Tag type is determined during production. To ensure a specific tag, please indicate your preference in advance; we will prioritize its development.
Synonyms
aroK; BT_3393Shikimate kinase; SK; EC 2.7.1.71
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-175
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacteroides thetaiotaomicron (strain ATCC 29148 / DSM 2079 / NCTC 10582 / E50 / VPI-5482)
Target Names
aroK
Target Protein Sequence
MVRIFLTGYM GAGKTTLGKA FARKLNVPFI DLDWYIEERF HKTVGELFTE RGEAGFRELE RNMLHEVAEF ENVVISTGGG APCFYDNMEF MNRTGKTVFL NVHPDVLFRR LRIAKQQRPI LQGKEDDELM DFIIQALEKR APFYTQAQYI FNADELEDRW QIESSVQRLQ ELLEL
Uniprot No.

Target Background

Function
This enzyme catalyzes the specific phosphorylation of the 3-hydroxyl group of shikimic acid using ATP as a co-substrate.
Database Links

KEGG: bth:BT_3393

STRING: 226186.BT_3393

Protein Families
Shikimate kinase family
Subcellular Location
Cytoplasm.

Q&A

What is the biological function of shikimate kinase in Bacteroides thetaiotaomicron?

Shikimate kinase (EC 2.7.1.71) catalyzes the specific phosphorylation of the 3-hydroxyl group of shikimic acid using ATP as a cosubstrate . This reaction represents the fifth step in the shikimate pathway, which is essential for aromatic amino acid biosynthesis in bacteria, including Bacteroides thetaiotaomicron . The enzyme plays a critical role in linking carbohydrate metabolism to the biosynthesis of chorismate, which serves as a precursor for aromatic amino acids and many other aromatic compounds . Since the shikimate pathway is present in microorganisms and plants but absent in mammals, B. thetaiotaomicron shikimate kinase represents a potential target for antimicrobial development with minimal host toxicity .

Why is recombinant expression of B. thetaiotaomicron shikimate kinase important for research?

Recombinant expression provides several key advantages for studying B. thetaiotaomicron shikimate kinase. First, it enables the production of sufficient quantities of pure enzyme for structural and functional characterization . Similar to other recombinant proteins from B. thetaiotaomicron, the aroK gene can be cloned and expressed in E. coli expression systems with affinity tags (such as His-tags) for simplified purification . This approach facilitates detailed enzymatic studies, crystallography experiments, and inhibitor screening that would be difficult with native protein preparations. Additionally, recombinant expression allows for site-directed mutagenesis to investigate the roles of specific amino acid residues in catalysis and substrate binding, contributing to mechanistic understanding of the enzyme and rational drug design efforts .

How does the structure of shikimate kinase relate to its function?

Shikimate kinases typically display a three-layer alpha/beta fold consisting of a central sheet of five parallel β-strands flanked by alpha-helices . Based on structural studies of other bacterial shikimate kinases, the enzyme undergoes an induced-fit conformational change from an open to a closed form upon substrate binding . The binding of shikimate typically occurs above a short 3₁₀ helix formed by a strictly conserved motif (GGGXV) . Several highly conserved charged residues, including those equivalent to Asp33, Arg57, and Arg132 in Helicobacter pylori shikimate kinase, are likely involved in shikimate binding and catalysis . These structural features enable the precise positioning of the 3-hydroxyl group of shikimate for phosphoryl transfer from ATP, resulting in the specific catalytic activity of the enzyme .

What is the recommended protocol for expressing and purifying recombinant B. thetaiotaomicron shikimate kinase?

Based on established protocols for similar recombinant proteins, B. thetaiotaomicron shikimate kinase can be efficiently expressed and purified using the following method:

  • Cloning and Expression Vector Construction:

    • Clone the aroK gene from B. thetaiotaomicron genomic DNA

    • Insert into an expression vector (e.g., pET or pQE series) with a C-terminal 6×His-tag

    • Transform into an E. coli expression strain such as BL21(DE3)

  • Protein Expression:

    • Grow transformed E. coli in LB medium with appropriate antibiotics at 37°C until mid-log phase

    • Induce expression with 0.5 mM IPTG at 16°C overnight to enhance protein solubility

    • Harvest cells by centrifugation and resuspend in lysis buffer (typically 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole)

  • Purification:

    • Lyse cells by sonication or French press

    • Clarify lysate by centrifugation at 15,000 × g for 30 minutes

    • Purify using immobilized nickel ion chromatography

    • Further purify by size exclusion chromatography using Superdex-75

    • Assess purity by SDS-PAGE

  • Storage:

    • Store in buffer containing 20 mM Tris-HCl pH 7.5, 100 mM NaCl at -80°C

    • Use a manual defrost freezer and avoid repeated freeze-thaw cycles

This protocol typically yields 20-30 mg of pure protein per liter of culture, suitable for enzymatic assays and structural studies.

How can the enzymatic activity of recombinant B. thetaiotaomicron shikimate kinase be measured?

The enzyme activity can be measured using a coupled spectrophotometric assay that links ADP production to NADH oxidation:

Materials required:

  • 100 mM Tris-HCl-KOH buffer, pH 7.5

  • 50 mM KCl

  • 5 mM MgCl₂

  • Shikimic acid (variable concentration for kinetic studies)

  • ATP (variable concentration for kinetic studies)

  • 1 mM phosphoenolpyruvate

  • 0.1 mM NADH

  • Pyruvate kinase (2.5 units/ml)

  • Lactate dehydrogenase (2.7 units/ml)

  • Purified recombinant B. thetaiotaomicron shikimate kinase

  • UV-visible spectrophotometer

Protocol:

  • Prepare reaction mixture containing all components except shikimate kinase

  • Establish baseline at 340 nm

  • Add shikimate kinase to initiate reaction

  • Monitor decrease in absorbance at 340 nm (ε = 6,200 M⁻¹ cm⁻¹)

  • Calculate initial velocity from the linear portion of the progress curve

For kinetic parameter determination:

  • For K₍ₘ₎ (ATP): Maintain shikimate at saturating concentration (e.g., 1.6 mM) and vary ATP (0.001-2.5 mM)

  • For K₍ₘ₎ (shikimate): Maintain ATP at saturating concentration (e.g., 2.5 mM) and vary shikimate (0.01-1.6 mM)

  • Fit data to Michaelis-Menten equation using nonlinear regression

Expected kinetic parameters for bacterial shikimate kinases typically fall within these ranges:

  • K₍ₘ₎ (ATP): 50-200 μM

  • K₍ₘ₎ (shikimate): 60-400 μM

  • k₍cat₎: 20-60 s⁻¹

How can structural data be used to design selective inhibitors of B. thetaiotaomicron shikimate kinase?

Designing selective inhibitors requires detailed structural information about the active site and substrate binding pocket of B. thetaiotaomicron shikimate kinase. This approach involves several steps:

  • Structural Determination:

    • Obtain crystal structures of the enzyme in both apo form and in complex with shikimate and/or ATP

    • Identify key binding interactions, particularly those involving the conserved motifs and residues

  • Comparative Analysis:

    • Compare with structures from other organisms to identify unique features

    • Focus on residues interacting with shikimate, including those equivalent to Asp33, Arg57, and Arg132 in H. pylori shikimate kinase

  • Structure-Based Design:

    • Utilize the induced-fit movement from open to closed form

    • Target the shikimate-binding site above the conserved GGGXV motif

    • Consider transition-state analogs that mimic the phosphoryl transfer reaction

  • Molecular Docking and Virtual Screening:

    • Employ computational methods to screen virtual libraries

    • Prioritize compounds that interact with conserved residues unique to bacterial enzymes

  • Validation:

    • Test predicted inhibitors using the enzymatic assay described in section 2.2

    • Determine inhibition constants and mechanisms (competitive, non-competitive, etc.)

    • Verify selectivity against mammalian kinases

This rational drug design approach has been successful for other shikimate pathway enzymes and holds promise for developing selective antimicrobials targeting B. thetaiotaomicron.

What is the role of B. thetaiotaomicron shikimate kinase in gut microbiome interactions?

B. thetaiotaomicron is a prominent member of the human gut microbiome with significant metabolic capabilities. Its shikimate kinase plays several important roles in microbiome interactions:

  • Essential Metabolism:

    • Provides aromatic amino acids that cannot be synthesized by the human host

    • Supports production of secondary metabolites derived from the shikimate pathway

  • Interspecies Competition:

    • Gives B. thetaiotaomicron an advantage in nutrient-limited conditions where aromatic amino acids are scarce

    • May affect community composition through differential growth capabilities

  • Host-Microbe Interactions:

    • Products of the shikimate pathway may serve as signaling molecules in host-microbe communication

    • Metabolites derived from this pathway can influence host immune responses

  • Dietary Interactions:

    • Dietary polyphenols may interact with or modulate the shikimate pathway

    • Plant-derived compounds may serve as alternative substrates or inhibitors

Research methodologies to study these interactions include:

  • Comparative genomics to analyze aroK conservation across gut bacterial species

  • Metabolomic analysis of shikimate pathway intermediates in gnotobiotic mouse models

  • Co-culture experiments to assess interspecies competition

  • Transcriptomic studies to determine regulation under different nutritional conditions

What are the key differences between shikimate kinase isoenzymes from different bacterial species?

Understanding the differences between shikimate kinase isoenzymes provides insights into evolutionary adaptations and helps in designing species-specific inhibitors:

ParameterE. coli AroKE. coli AroLH. pylori SKM. jannaschii SKB. thetaiotaomicron SK*
K₍ₘ₎ (shikimate) (μM)~310~20060 ± 8414 ± 3360-200*
K₍ₘ₎ (ATP) (μM)Not reportedNot reported101 ± 1748 ± 450-150*
V₍max₎ (μmol/min/mg)Not reportedNot reported22-26Not reported20-30*
Sequence identity with E. coli AroK100%~30%LowVery lowModerate*
Structural featuresStandardStandardInduced-fit upon bindingGHMP-kinase family relatedPredicted standard fold*

*Values for B. thetaiotaomicron SK are estimated based on related bacterial shikimate kinases

Key differences among these enzymes include:

  • Substrate Affinity:

    • H. pylori SK shows higher affinity for shikimate than E. coli enzymes

    • Archaeal SKs (e.g., M. jannaschii) show distinct kinetic parameters

  • Structural Variations:

    • Most bacterial SKs share the core alpha/beta fold

    • Archaeal SKs are distantly related to homoserine kinases of the GHMP-kinase superfamily

    • Variations in the shikimate-binding domain and LID region affect substrate specificity

  • Regulation:

    • Some species contain two isoenzymes (AroK and AroL) with different regulatory properties

    • Single-isoenzyme species may have different regulatory mechanisms

These differences can be exploited for species-specific targeting in antimicrobial development.

What are common challenges in expressing active recombinant B. thetaiotaomicron shikimate kinase and how can they be overcome?

Researchers often encounter several challenges when expressing recombinant B. thetaiotaomicron shikimate kinase:

  • Poor Solubility:

    • Problem: Formation of inclusion bodies

    • Solution:

      • Reduce induction temperature to 16°C

      • Use solubility-enhancing fusion tags like SUMO or MBP

      • Optimize expression by testing different E. coli strains (BL21, Rosetta, Arctic Express)

      • Consider codon optimization for E. coli expression

  • Low Activity:

    • Problem: Enzyme expresses but shows minimal activity

    • Solution:

      • Ensure proper folding by adding chaperones (co-express GroEL/ES)

      • Verify buffer conditions, particularly Mg²⁺ concentration

      • Check for inhibitory contaminants in purification

      • Consider tag position (N- versus C-terminal) effects on activity

  • Protein Instability:

    • Problem: Rapid loss of activity during storage

    • Solution:

      • Add stabilizing agents (glycerol 10-20%, reducing agents)

      • Optimize buffer composition and pH

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

      • Consider addition of substrate analogs for stabilization

  • Heterogeneity:

    • Problem: Multiple forms or degradation products

    • Solution:

      • Add protease inhibitors during purification

      • Optimize purification protocol with additional chromatography steps

      • Consider site-directed mutagenesis of sensitive sites

      • Analyze by mass spectrometry to identify modifications

These methodological approaches have proven effective for other recombinant proteins from B. thetaiotaomicron and should be applicable to shikimate kinase .

How can researchers differentiate between enzymatic activity of shikimate kinase and other ATP-utilizing enzymes in crude preparations?

Distinguishing shikimate kinase activity from other ATP-utilizing enzymes requires specific methodological approaches:

  • Substrate Specificity:

    • Measure activity with and without shikimate

    • True shikimate kinase activity should be strictly dependent on shikimate presence

    • Control reactions with structurally similar compounds can identify non-specific kinases

  • Inhibition Studies:

    • Use known shikimate kinase inhibitors or substrate analogs

    • Compare inhibition profiles with those of characterized shikimate kinases

    • Differential inhibition patterns can distinguish between enzyme types

  • Immunological Methods:

    • Use antibodies raised against recombinant shikimate kinase

    • Perform immunodepletion studies to remove specific enzyme

    • Quantify activity before and after immunodepletion

  • Chromatographic Separation:

    • Use ion exchange or size exclusion chromatography to separate enzymes

    • Analyze shikimate kinase activity in different fractions

    • Compare with western blot analysis of fractions

  • Product Analysis:

    • Use HPLC or mass spectrometry to directly detect shikimate 3-phosphate formation

    • Confirm product identity with authentic standards

    • This approach provides direct evidence of specific activity

These methods can be combined for robust differentiation between shikimate kinase and other ATP-utilizing enzymes in complex biological samples.

How does shikimate kinase essentiality vary across different bacterial pathogens, and what are the implications for antimicrobial development?

The essentiality of shikimate kinase varies among bacterial species, which has important implications for antimicrobial development:

  • Comparative Essentiality:

    • Acinetobacter baumannii: aroK gene is essential for growth and survival during host infection

    • Similar essentiality observed in other pathogens lacking alternative aromatic amino acid acquisition pathways

    • Some bacteria with robust transport systems may show reduced essentiality

  • Validation Methods:

    • Gene knockout studies in different infection models

    • Conditional expression systems to control gene expression

    • Chemical genetics approaches using targeted inhibitors

    • Comparative genomics to identify natural aroK deletion mutants

  • Implications for Drug Development:

    • High-priority targets should focus on pathogens where aroK is most essential

    • Combination therapies may be needed for species with alternative pathways

    • Species-specific structural differences can be exploited for selective targeting

    • Broad-spectrum potential exists for conserved binding site features

  • Research Methodology:

    • In vivo infection models are crucial for validating essentiality

    • Transcriptomic studies under different growth conditions

    • Metabolic flux analysis to quantify pathway importance

    • Comparative structural biology to identify exploitable differences

This comparative approach helps prioritize shikimate kinase as an antimicrobial target against specific pathogens where its essentiality is well-established.

What emerging technologies are advancing our understanding of shikimate kinase enzymology?

Several cutting-edge technologies are revolutionizing shikimate kinase research:

  • Cryo-Electron Microscopy (Cryo-EM):

    • Enables visualization of enzyme conformational dynamics

    • Captures intermediate states during catalysis

    • Provides insights into protein-protein interactions within metabolic complexes

    • Methodology: Sample vitrification, high-resolution imaging, computational reconstruction

  • Time-Resolved X-ray Crystallography:

    • Captures structural changes during catalytic cycle

    • Reveals transient binding states not visible in static structures

    • Methodology: Microcrystals, X-ray free-electron lasers, pump-probe experiments

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):

    • Maps conformational changes upon substrate binding

    • Identifies regions of altered solvent accessibility during catalysis

    • Methodology: Controlled H/D exchange, proteolysis, LC-MS/MS analysis

  • Single-Molecule Enzymology:

    • Observes individual enzyme molecules rather than ensemble averages

    • Reveals catalytic heterogeneity and rare conformational states

    • Methodology: Fluorescence resonance energy transfer (FRET), total internal reflection fluorescence (TIRF)

  • Computational Methods:

    • Molecular dynamics simulations of full catalytic cycle

    • Quantum mechanics/molecular mechanics (QM/MM) for transition state analysis

    • Machine learning approaches for inhibitor discovery

    • Methodology: High-performance computing, specialized software packages

These technologies are providing unprecedented insights into the structural dynamics and catalytic mechanisms of shikimate kinase, informing both basic understanding and applied drug discovery efforts.

How might genetic engineering of B. thetaiotaomicron shikimate kinase enhance aromatic amino acid production for synthetic biology applications?

Genetic engineering of B. thetaiotaomicron shikimate kinase offers several strategies for enhancing aromatic amino acid production:

  • Enzyme Engineering Approaches:

    • Rational design to reduce feedback inhibition

    • Directed evolution to enhance catalytic efficiency

    • Protein fusion strategies to create metabolic channeling

    • Methodology: Site-directed mutagenesis, error-prone PCR, high-throughput screening

  • Metabolic Engineering Strategies:

    • Overexpression of wild-type or engineered aroK

    • Deletion of competing pathways (similar to E. coli aroL/aroK deletions)

    • Co-expression with other rate-limiting enzymes in the pathway

    • Methodology: CRISPR-Cas9 genome editing, plasmid-based expression systems

  • Regulatory Engineering:

    • Promoter optimization for controlled expression

    • Removal of transcriptional repressors

    • Engineering of allosteric regulation

    • Methodology: Promoter libraries, transcription factor engineering

  • Process Optimization:

    • Identification of optimal carbon sources (e.g., sorbitol showed enhanced production in E. coli)

    • Fed-batch cultivation strategies

    • Bioprocess parameter optimization

    • Methodology: Bioreactor studies, metabolic flux analysis

Implementation of these approaches could significantly improve aromatic amino acid production in B. thetaiotaomicron, with potential applications in sustainable biochemical production.

What methodological approaches can elucidate the role of shikimate kinase in B. thetaiotaomicron's adaptation to the human gut environment?

Understanding the role of shikimate kinase in gut adaptation requires sophisticated methodological approaches:

  • In vivo Colonization Studies:

    • Gnotobiotic mouse models comparing wild-type and aroK mutants

    • Competitive colonization assays

    • Multi-omics analysis of colonized tissues

    • Methodology: Germ-free mouse facilities, high-throughput sequencing, metabolomics

  • Environmental Response Analysis:

    • Transcriptomic profiling under gut-relevant conditions

    • Proteomics to assess enzyme levels in different microenvironments

    • Metabolic profiling to track shikimate pathway flux

    • Methodology: RNA-seq, mass spectrometry, stable isotope labeling

  • Interaction Studies:

    • Co-culture experiments with other gut microbes

    • Host cell-microbe interaction assays

    • Effect of dietary components on aroK expression

    • Methodology: Fluorescence microscopy, transwell systems, metabolite analysis

  • Evolutionary Analysis:

    • Comparative genomics across B. thetaiotaomicron strains

    • Analysis of aroK sequence variation in human gut isolates

    • Identification of selective pressures on the shikimate pathway

    • Methodology: Whole genome sequencing, phylogenetic analysis, population genetics

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.