Recombinant Bacillus cereus Urease accessory protein UreD (ureD)

Shipped with Ice Packs
In Stock

Description

UreD Function and Characteristics

UreD, along with UreF and UreG, forms a complex that acts as a GTP-hydrolysis-dependent molecular chaperone . This complex is crucial for activating the urease apoprotein by assisting in the assembly of the urease metallocenter . Specifically, it helps in the insertion of two nickel ions (Ni2+) into the active site of the urease apoprotein (UreABC) .

UreD in Klebsiella aerogenes

Research on UreD in Klebsiella aerogenes provides further insight into its function. In K. aerogenes, four accessory proteins—UreD, UreE, UreF, and UreG—are required for the effective delivery and incorporation of two Ni2+ ions into the urease apoprotein (UreABC) .

A translational fusion between maltose-binding protein (MBP) and UreD was found to be soluble in Escherichia coli cell extracts and capable of complementing a ΔureD-urease cluster in this host microorganism . The purified MBP-UreD formed a large multimer (>670 kDa) that bound approximately 2.5 Ni2+ ions (Kd of ∼50 μM) per UreD protomer, as determined by equilibrium dialysis measurements . Zinc ions (Zn2+) competed directly with a 10-fold higher affinity (∼4 Zn2+ ions per protomer; Kd of 5 μM) for the Ni2+ binding sites .

MBP pulldown experiments showed that the UreD domain of MBP-UreD formed in vivo complexes with UreF, UreG, UreF plus UreG, or UreABC when these proteins were overproduced in the same E. coli cells . Additionally, a UreABC-(MBP-UreD)-UreFG complex was observed in cells producing all urease components .

Urease Activity and Nitrogen Metabolism in Bacillus cereus

A study examined the prevalence of urease genes and activity among 49 environmental, food, and clinical B. cereus isolates . Ten strains were found to have urease genes, with eight of these strains capable of growing on urea as the sole nitrogen source . Interestingly, two urease-positive strains, including the sequenced strain ATCC 10987, could not utilize urea for growth, despite producing active urease, due to their inability to use ammonium as a nitrogen source . The effect of urea hydrolysis on acid stress resistance was also assessed among the ureolytic B. cereus strains, but none showed increased fitness under acidic conditions or enhanced acid shock survival in the presence of urea .

Role of EntD in Bacillus cereus

EntD, another protein in Bacillus cereus, has been identified as a crucial protein affecting cell wall structure and various cellular processes . While not directly related to UreD, understanding EntD can provide a broader context of protein functions in B. cereus.

EntD is an exoprotein with SH3 domains involved in protein-protein interactions and a 3D domain for cell wall binding . Disruption of entD significantly decreased the growth rate and acetate overflow in B. cereus without changing the final biomass . A ΔentD mutant showed lower accumulation of flagellum components and was non-flagellated, leading to reduced swimming motility but increased biofilm formation . EntD appears to play a vital role in maintaining cell wall structure, and its deficiency activates a cellular response involving cell wall remodeling, metabolism, and virulence regulators .

Product Specs

Form
Lyophilized powder Note: We will prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them during order placement. We will accommodate your request to the best of our ability.
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 otherwise specified. 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%, which can serve as a reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein. 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 will be determined during the manufacturing process. The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
ureD; BCE_3658; Urease accessory protein UreD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-273
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacillus cereus (strain ATCC 10987 / NRS 248)
Target Names
ureD
Target Protein Sequence
MKSPTGVLNI DVMEKRDKTV PIKVYHKDAL KVTQPIYLDK YGRAYYYIMN SGGGYLKGDF YSININVGKD AKTYITSQSA TKVYKTPNSY ALQELNFYIG QNAAMEYLPD PLIMYKDAAY KQKTNIYMQN NSTLILCDSV TPGWSPNMEK FTYQYFDSLT KIYMENKLVV YDHLLLNPFK ESLDQMGILN QYSHYGTFIV INENVTNDLI AALKTSFSNT NNMKIGIAST TCKGFVIRIL SHNTEDMESI FFQCHRFVRE NCLHEELTSY RKY
Uniprot No.

Target Background

Function

Essential for urease maturation through the functional incorporation of the urease nickel metallocenter.

Database Links

KEGG: bca:BCE_3658

Protein Families
UreD family
Subcellular Location
Cytoplasm.

Q&A

What is the functional role of Urease accessory protein UreD in Bacillus cereus?

UreD is one of several accessory proteins essential for the assembly and activation of the urease metalloenzyme complex in bacteria, including B. cereus. The protein serves as a molecular chaperone that facilitates the incorporation of nickel ions into the urease active site. In the B. cereus group, UreD likely participates in a complex with other accessory proteins (UreF, UreG) to form a molecular scaffold that interacts with the urease apoprotein prior to nickel insertion and enzyme activation.

Methodologically, researchers can assess UreD function through complementation studies in urease-deficient strains. Expression of recombinant UreD in these strains, followed by measurement of urease activity using colorimetric assays based on ammonium production, can confirm the protein's role in enzyme activation. Additionally, pull-down assays and co-immunoprecipitation can identify protein interaction partners within the urease maturation pathway.

Which expression systems yield optimal production of recombinant B. cereus UreD?

For B. cereus group proteins, several expression systems have demonstrated effectiveness. Based on approaches similar to those used for other B. cereus proteins, the following systems may be considered:

  • E. coli-based expression: BL21(DE3) strains with pET vector systems often provide high yields when expressing B. cereus proteins. The high-throughput screening method utilizing "heat-shock method at 42°C for 90 s and cultured in LB medium containing 50 μg/mL kanamycin at 37°C for 12 h" has proven successful for B. cereus group recombinant proteins .

  • Bacillus-based expression: Using B. subtilis as a host may improve proper folding of B. cereus proteins due to evolutionary similarities. For instance, successful expression of B. subtilis glucose dehydrogenase (BsGDH) in B. cereus suggests compatibility within the genus .

Expression protocols might include induction at lower temperatures (22°C) with moderate IPTG concentrations (0.5 mM) to enhance solubility, as demonstrated for other B. cereus group proteins .

How should researchers optimize purification protocols for recombinant B. cereus UreD?

Purification of recombinant UreD from B. cereus requires careful consideration of protein properties to maintain structural integrity and function. A multi-step purification approach is recommended:

  • Cell lysis optimization: Gentle lysis techniques using lysozyme (0.5 mg/mL) combined with freeze-thaw cycles have proven effective for B. cereus group proteins .

  • Affinity chromatography: His-tagged UreD can be purified using nickel affinity chromatography with imidazole gradient elution.

  • Size exclusion chromatography: This secondary purification step helps separate monomeric UreD from aggregates and other contaminants.

Protein stability should be monitored throughout purification using techniques such as circular dichroism spectroscopy and thermal shift assays. Buffer composition should be optimized for pH and salt concentration to maintain native conformation.

What molecular techniques can verify successful cloning of B. cereus ureD?

Verification of recombinant B. cereus ureD cloning requires multiple molecular techniques:

  • Restriction enzyme analysis: Digest plasmid DNA with appropriate restriction enzymes to confirm insert size.

  • PCR verification: Design primers specific to the ureD gene sequence and vector backbone junctions.

  • DNA sequencing: Complete sequence verification is essential, particularly for expression constructs. Commercial sequencing services like "Sangon Biotech" have been successfully used for verification of B. cereus group recombinant genes .

  • Expression verification: Small-scale expression followed by Western blot analysis using antibodies against the affinity tag or the UreD protein directly.

For PCR-based verification, typical reaction conditions include similar annealing temperatures to those used for other B. cereus group genes (56-59°C) .

How does UreD from B. cereus compare functionally to UreD proteins from other members of the B. cereus group?

The B. cereus group comprises closely related species including B. anthracis, B. thuringiensis, and B. mycoides . Despite their phenotypic differences, these species show remarkable genomic similarity, suggesting potential conservation of urease accessory proteins across the group.

Comparative functional analysis requires:

  • Sequence alignment: Multiple sequence alignment of UreD proteins from various B. cereus group species to identify conserved domains and species-specific variations.

  • Cross-complementation studies: Expressing UreD from different species in a common urease-deficient host to assess functional conservation.

  • Protein-protein interaction mapping: Investigating whether UreD from B. cereus interacts with urease components from related species through techniques like bacterial two-hybrid assays.

Given the close phylogenetic relationship revealed by multilocus sequence typing (MLST) studies of the B. cereus group , researchers should consider the evolutionary context when interpreting functional differences between UreD proteins.

What site-directed mutagenesis approaches are most effective for studying B. cereus UreD functional domains?

Site-directed mutagenesis of B. cereus UreD can reveal critical functional residues and domains. Based on successful approaches with other B. cereus proteins, the following methodologies are recommended:

  • Saturation mutagenesis: This approach, as demonstrated for BsGDH in B. cereus, can systematically replace specific amino acid residues to identify those critical for protein function .

  • Alanine-scanning mutagenesis: Systematically replacing residues with alanine to identify those essential for protein-protein interactions or catalytic function.

  • Conservation-guided mutagenesis: Focus on "low-conserved residues" in the protein's secondary structure, as this approach successfully identified critical functional residues in other B. cereus proteins .

The mutagenesis protocol can follow established methods, including:

  • PCR-based mutagenesis using complementary primers containing the desired mutation

  • DpnI digestion to remove template DNA

  • Transformation into high-efficiency competent cells

Analysis of mutant phenotypes should include assessment of urease activation efficiency, protein stability, and interaction with other urease components.

How can structural biology techniques advance understanding of B. cereus UreD interaction networks?

Structural characterization of B. cereus UreD and its interaction partners requires sophisticated approaches:

  • X-ray crystallography: For high-resolution structure determination of purified UreD alone or in complex with interacting partners.

  • Cryo-electron microscopy: Particularly valuable for capturing the UreD-urease complex assembly process.

  • Molecular dynamics simulations: Computational approaches can reveal conformational changes in UreD during complex formation. Root Mean Square Fluctuation (RMSF) analysis, as applied to BsGDH mutants, can identify regions of increased flexibility that may be important for function .

  • Hydrogen-deuterium exchange mass spectrometry: To map interaction interfaces between UreD and other urease accessory proteins.

Research in similar B. cereus proteins has shown that molecular docking and dynamics simulations can successfully identify critical structural features like the "triangular region" that enhances substrate affinity in BsGDH .

What is the relationship between B. cereus UreD function and pathogenicity?

The potential role of UreD in B. cereus pathogenicity remains an important research question, especially considering the diverse pathogenic properties within the B. cereus group:

  • Comparative genomic analysis: Examine ureD presence, absence, and variation across pathogenic and non-pathogenic B. cereus strains.

  • Virulence models: Compare urease activity and pathogenicity in wild-type versus ureD knockout strains in appropriate infection models.

  • Transcriptomic analysis: Investigate ureD expression patterns under conditions mimicking host environments.

The B. cereus group exhibits "highly divergent pathogenic properties" despite close genetic relationships . Some B. cereus strains cause food poisoning and soft tissue infections, while others may be primarily environmental . Understanding UreD's potential contribution to these diverse phenotypes requires careful experimental design and controls.

How can advanced recombinant protein engineering improve B. cereus UreD stability and functionality?

Protein engineering approaches can enhance the properties of recombinant B. cereus UreD for research applications:

  • Fusion protein design: Creating fusion constructs with solubility-enhancing tags like MBP (maltose-binding protein) or SUMO.

  • Disulfide engineering: Strategic introduction of disulfide bonds to enhance thermostability.

  • Directed evolution: Development of UreD variants with enhanced stability or activity through iterative rounds of mutation and selection.

Drawing from the success with BsGDH mutants in B. cereus, researchers can apply similar directed evolution approaches that resulted in "5.66 and 11.38 times greater" activity compared to wild-type enzymes . The three-round saturation mutagenesis strategy targeting low-conserved residues provides a methodological template for UreD engineering .

What controls are essential when studying recombinant B. cereus UreD function?

Rigorous experimental controls are critical for UreD functional studies:

  • Negative controls:

    • Empty vector expression in the same host

    • Catalytically inactive UreD mutant (based on conserved residues)

    • Urease apoprotein without accessory proteins

  • Positive controls:

    • Well-characterized UreD from model organisms like Klebsiella aerogenes

    • Wild-type B. cereus containing native urease system

  • Specificity controls:

    • Related accessory proteins (UreF, UreG) to confirm specific effects

    • UreD from closely related B. cereus group species like B. anthracis or B. thuringiensis

Experimental design should account for the clonal population structure of the B. cereus group revealed by multilocus sequence typing studies , ensuring bacterial strains are properly characterized phylogenetically.

What are the optimal conditions for assessing B. cereus UreD-mediated urease activation?

Assessing UreD function requires careful optimization of reaction conditions:

ParameterRange to TestNotes
pH6.0-8.0Test at 0.5 pH unit intervals
Temperature25-37°COptimal growth temperature for B. cereus is typically 30-35°C
Nickel concentration0-100 μMEssential cofactor for urease activity
Incubation time15-120 minMonitor time course of activation
Reducing agents0-10 mM DTT or β-MEMay affect cysteine residues in UreD

Urease activity can be quantified using standard assays that measure ammonia production from urea hydrolysis, with activity typically expressed in U/mL or U/mg protein, similar to how antibacterial activity was quantified for modified B. cereus strains .

How can researchers address low solubility of recombinant B. cereus UreD?

Protein solubility challenges are common with recombinant expression. For B. cereus UreD, consider:

  • Expression optimization:

    • Lower induction temperature (16-22°C), similar to the 22°C used for other B. cereus recombinant proteins

    • Reduced IPTG concentration (0.1-0.5 mM)

    • Co-expression with molecular chaperones

  • Buffer optimization:

    • Screen additives: glycerol (5-10%), salt concentration (100-500 mM NaCl)

    • Test different pH conditions (pH 6.5-8.5)

    • Add stabilizing agents like arginine or trehalose

  • Protein engineering:

    • Truncation constructs to remove potentially aggregation-prone regions

    • Fusion to solubility-enhancing tags

Use techniques like dynamic light scattering to monitor aggregation state during optimization.

What strategies help overcome challenges in detecting protein-protein interactions involving B. cereus UreD?

Detecting interactions between UreD and other urease components can be challenging. Recommended approaches include:

  • In vitro methods:

    • Pull-down assays with differentially tagged proteins

    • Surface plasmon resonance to measure binding kinetics

    • Isothermal titration calorimetry for thermodynamic characterization

  • In vivo methods:

    • Bacterial two-hybrid systems adapted for Bacillus species

    • Fluorescence resonance energy transfer (FRET) with fluorescently tagged proteins

    • Co-immunoprecipitation from bacterial lysates

  • Crosslinking strategies:

    • Chemical crosslinkers of various arm lengths

    • Photo-activatable crosslinkers for capturing transient interactions

When interpreting protein interaction data, consider that the B. cereus group shows "a generally clonal structure to the population" , which may influence conservation of interaction networks across strains.

How can mass spectrometry elucidate post-translational modifications of B. cereus UreD?

Mass spectrometry provides powerful tools for characterizing UreD post-translational modifications (PTMs) that may regulate its function:

  • Sample preparation:

    • Purify recombinant UreD to high homogeneity

    • Use multiple proteases for digestion to maximize sequence coverage

    • Enrich for specific PTMs if targeting particular modifications

  • MS techniques:

    • LC-MS/MS for peptide mapping and modification identification

    • Top-down proteomics for intact protein analysis

    • Targeted MS methods for quantifying specific modifications

  • Data analysis:

    • Search against comprehensive PTM databases

    • Use appropriate statistical tools to distinguish true modifications from artifacts

    • Consider comparative analysis between active and inactive UreD states

This approach can identify modifications similar to those revealed in other B. cereus proteins that affect their functional properties.

What computational approaches best predict B. cereus UreD structure and function?

Computational methods offer valuable insights when experimental structural data is limited:

  • Homology modeling:

    • Identify suitable templates from structurally characterized UreD proteins

    • Generate multiple models and validate using quality assessment tools

    • Refine models through molecular dynamics simulations

  • Molecular docking:

    • Predict UreD interactions with other urease components

    • Identify potential binding sites for cofactors or inhibitors

    • Validate predictions through mutagenesis studies

  • Sequence-based predictions:

    • Identify conserved domains through multiple sequence alignment

    • Predict disorder regions that may facilitate protein-protein interactions

    • Use coevolution analysis to identify residue pairs that may interact

Molecular docking approaches successfully identified critical structural features in other B. cereus proteins, such as the "triangular region formed by residues Gly94, Gly14, and Ile191" in BsGDH , suggesting similar approaches would be valuable for UreD analysis.

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.