Recombinant Bacillus subtilis Uncharacterized protein ykoA (ykoA)

Shipped with Ice Packs
In Stock

Description

Production and Expression Systems

Recombinant ykoA is produced in Escherichia coli using standard molecular biology techniques. Key production parameters include:

ParameterValue
Host OrganismE. coli
Expression SystemRecombinant protein production with N-terminal His-tag
Purification MethodAffinity chromatography (via His-tag)
Purity>90% (SDS-PAGE)
YieldNot explicitly reported (commercial products available in 50–100 µg quantities)

The choice of E. coli as the host reflects its cost-effectiveness and scalability for recombinant protein production, though B. subtilis itself is a widely used host for secretion-competent proteins .

Physical and Biochemical Properties

Critical handling and storage protocols ensure protein stability:

ParameterValue
Storage Conditions-20°C or -80°C (lyophilized powder)
Reconstitution BufferDeionized sterile water (with optional 5–50% glycerol)
StabilityAvoid repeated freeze-thaw cycles; aliquot for multiple use

The protein is supplied in Tris/PBS-based buffer with 6% trehalose or 50% glycerol to prevent denaturation .

Research Gaps and Future Directions

Despite its availability, ykoA remains functionally uncharacterized. Current research focuses on:

  • Structural Elucidation: Crystallization or cryo-EM studies to identify potential ligand-binding sites or catalytic motifs.

  • Functional Annotation: Bioinformatics approaches (e.g., homology modeling, gene knockout studies in B. subtilis) to infer biological roles.

  • Biotechnological Applications: Exploration in protein engineering, surface display, or biocatalysis, leveraging B. subtilis’s secretion capabilities .

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have specific requirements for the format, please indicate them in your order. We will prepare the product according to your needs.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery times.
Note: All our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance, as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents settle to the bottom. Please reconstitute the protein in deionized sterile 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 default final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by several factors, including storage conditions, buffer ingredients, storage temperature, and the protein's inherent stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type is determined during production. If you have a specific tag type requirement, please inform us, and we will prioritize developing the specified tag.
Synonyms
ykoA; BSU14420; Uncharacterized protein YkoA
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-89
Protein Length
full length protein
Species
Bacillus subtilis (strain 168)
Target Names
ykoA
Target Protein Sequence
MRLLTLTEYCLLIFFTGFYLAVTGFTAKDIGLYIGIALIYIFSHIFSKRLLEKRGKENKQ VHLFFSVLAIIGSVFITVLCIALVASFSK
Uniprot No.

Target Background

Database Links
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is Bacillus subtilis and why is it important for recombinant protein expression?

Bacillus subtilis is a rod-shaped, Gram-positive bacterium primarily found in soil, air, and decomposing plant matter. Initially classified as Vibrio subtilis in 1835, it was reclassified by Ferdinand Julius Cohn in 1872 . This organism has become a cornerstone in biotechnology and research for several key reasons. It possesses high stress resistance, exhibits non-toxic properties, demonstrates low codon preference, and grows rapidly . Most significantly, B. subtilis contains at least three distinct protein secretion pathways alongside abundant molecular chaperones, which collectively enhance its expression capabilities and compatibility with various recombinant proteins . These characteristics make it especially valuable for the expression of uncharacterized proteins like ykoA.

The bacterium's ability to form endospores allows it to survive extreme environmental conditions, making it robust for laboratory cultivation . Furthermore, B. subtilis has emerged as one of the most thoroughly studied model organisms next to Escherichia coli, with a fully sequenced genome that enables precise genetic manipulation . Its genetic competence—the developmental state in which it actively takes up exogenous DNA—makes it particularly amenable to transformation and genetic engineering approaches .

What are uncharacterized proteins in B. subtilis and why study them?

Uncharacterized proteins like ykoA represent gene products whose functions, structures, and biological roles remain largely unknown despite their conservation in the bacterial genome. Studying these proteins is crucial for several reasons:

  • Completing the functional annotation of the B. subtilis genome

  • Discovering novel enzymatic activities with potential biotechnological applications

  • Understanding previously unrecognized biological pathways and regulatory networks

  • Identifying potential antimicrobial targets in related pathogenic species

  • Expanding our fundamental knowledge of bacterial physiology and metabolism

B. subtilis serves as a model organism for many important pathogens including Bacillus anthracis, Staphylococcus aureus, and Listeria monocytogenes . Therefore, characterizing proteins in B. subtilis often provides valuable insights into homologous proteins in these pathogenic bacteria, potentially leading to new therapeutic approaches.

What expression systems are typically used for recombinant B. subtilis uncharacterized proteins?

For the expression of uncharacterized B. subtilis proteins like ykoA, researchers typically utilize either homologous (within B. subtilis) or heterologous (often E. coli or yeast) expression systems . The choice depends on research objectives:

Homologous Expression in B. subtilis:

  • Provides native post-translational modifications and protein folding environment

  • Utilizes the bacterium's efficient protein secretion capabilities

  • Benefits from minimal endotoxin production, making purification simpler

  • Requires optimization of promoters, expression vectors, and signal peptides for efficient expression

Heterologous Expression in E. coli or Yeast:

  • E. coli offers high-yield expression and well-established protocols

  • Yeast systems provide eukaryotic processing capabilities when needed

  • Both systems have extensive genetic tool availability

  • Available as standardized commercial expression systems

The expression system selection should consider the protein's characteristics, required yield, downstream applications, and whether native conformation is essential for functional studies.

How can I optimize the expression of recombinant B. subtilis uncharacterized protein ykoA?

Optimizing expression of uncharacterized proteins like ykoA requires a systematic approach addressing multiple factors:

Promoter Selection and Optimization:
The choice of promoter significantly impacts expression levels. For B. subtilis expression systems, researchers should consider:

  • Constitutive promoters for continuous expression

  • Inducible promoters (e.g., IPTG-inducible, xylose-inducible) for controlled expression

  • Promoter strength matching the desired expression level

Vector System Optimization:

  • Select vectors with appropriate copy numbers

  • Ensure compatibility with the chosen promoter system

  • Consider vectors with suitable selection markers for the host strain

  • Evaluate vectors with optimal regulatory elements that increase protein synthesis and secretion

Signal Peptide Selection:
For secreted expression, the choice of signal peptide is crucial:

  • Test multiple signal peptides as their efficiency varies with different proteins

  • Consider Sec or Tat pathway-specific signal sequences based on protein folding requirements

  • Optimize the signal peptide sequence if necessary

Host Strain Selection:

  • Use protease-deficient strains to minimize protein degradation

  • Consider genome-minimized B. subtilis strains which have shown superior performance for difficult proteins

  • Evaluate strains with enhanced chaperone expression for complex proteins

Expression Conditions:

  • Optimize temperature, media composition, and induction parameters

  • Consider fed-batch cultures for higher cell densities and protein yields

  • Monitor growth rates and adjust induction timing accordingly

Codon Optimization:
Although B. subtilis has low codon preference compared to other expression systems, codon optimization may still improve expression of specific proteins with problematic codon usage .

What purification strategies are most effective for recombinant B. subtilis uncharacterized proteins?

Purification of uncharacterized proteins presents unique challenges due to limited information about their properties. A systematic approach includes:

Affinity Tag Selection:

  • Polyhistidine (His) tags remain the most versatile option

  • Fusion partners like GST, MBP, or SUMO can improve solubility

  • Consider C-terminal vs. N-terminal tag placement based on predicted protein structure

  • Avi-tag biotinylation systems enable highly specific purification through biotin-streptavidin interactions

Tag Removal Considerations:

  • Include protease cleavage sites if native protein is required

  • Test multiple proteases (TEV, PreScission, etc.) if tag removal proves difficult

  • Assess whether the tag affects protein function through comparative assays

Chromatography Strategy:

  • Initial capture: Affinity chromatography based on selected tag

  • Intermediate purification: Ion exchange chromatography based on predicted pI

  • Polishing: Size exclusion chromatography to remove aggregates and ensure homogeneity

Specific Challenges for Uncharacterized Proteins:

  • Conduct stability tests in various buffers to identify optimal purification conditions

  • Perform small-scale expression tests to assess solubility before scale-up

  • Consider on-column refolding strategies if the protein forms inclusion bodies

  • Implement high-throughput screening approaches to rapidly identify optimal conditions

Quality Control:

  • SDS-PAGE and Western blotting to confirm protein identity and purity

  • Mass spectrometry for accurate molecular weight determination

  • Circular dichroism to assess secondary structure integrity

  • Dynamic light scattering to evaluate homogeneity and aggregation state

What analytical techniques are most informative for initial characterization of uncharacterized B. subtilis proteins?

Initial characterization of uncharacterized proteins requires a multi-technique approach:

Structural Analysis:

  • Circular dichroism (CD) spectroscopy for secondary structure assessment

  • Thermal shift assays to determine protein stability

  • Limited proteolysis to identify stable domains

  • Small-angle X-ray scattering (SAXS) for low-resolution structural information

  • Crystallization screening if high-resolution structure is needed

Functional Analysis:

  • Bioinformatic analysis for domain identification and homology to characterized proteins

  • Enzymatic activity screening using substrate libraries

  • Protein-protein interaction studies (pull-downs, yeast two-hybrid, BLI, SPR)

  • Subcellular localization studies using fluorescent fusion proteins

  • Phenotypic analysis of knockout/overexpression strains

Biophysical Characterization:

  • Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) for oligomeric state determination

  • Isothermal titration calorimetry (ITC) for binding studies

  • Differential scanning calorimetry (DSC) for thermal stability analysis

  • Analytical ultracentrifugation for shape and association state information

How can genomic context analysis help in predicting the function of uncharacterized B. subtilis proteins like ykoA?

Genomic context analysis offers powerful insights into potential functions of uncharacterized proteins through examination of their genomic neighborhood and evolutionary patterns:

Operon Structure Analysis:

  • Genes within the same operon often function in related biological processes

  • Transcriptional regulation patterns can indicate functional relationships

  • Co-expression with characterized genes provides functional clues

Comparative Genomics Approaches:

  • Phylogenetic profiling to identify co-evolved gene clusters

  • Synteny analysis across related species to identify conserved genomic neighborhoods

  • Horizontal gene transfer analysis to identify recently acquired functions

Protein Domain Architecture:

  • Identification of recognized domains through bioinformatic tools

  • Domain fusion events that suggest functional associations

  • Analysis of conserved residues that may indicate active sites or binding interfaces

Implementation Strategy:

  • Extract the genomic context of the ykoA gene in B. subtilis

  • Identify homologs in related species using BLAST or similar tools

  • Compare genomic neighborhoods across multiple species

  • Look for conserved gene clusters or operons containing ykoA homologs

  • Analyze transcriptomic data to identify co-expressed genes

  • Integrate findings to develop testable hypotheses about function

This approach has proven particularly valuable for B. subtilis, as its genome has been subject to extensive analysis and annotation over decades of research .

What are the challenges in structural biology approaches for uncharacterized B. subtilis proteins?

Structural determination of uncharacterized proteins presents several specific challenges that require strategic approaches:

Expression and Purification Challenges:

  • Difficulty predicting optimal expression conditions without functional knowledge

  • Potential toxicity when overexpressed in host systems

  • Unstable proteins that degrade during purification

  • Formation of inclusion bodies requiring refolding strategies

Crystallization Barriers:

  • Intrinsically disordered regions hindering crystal formation

  • Conformational heterogeneity affecting crystal packing

  • Need for ligands or binding partners to stabilize active conformations

  • Limited prior knowledge to guide crystallization condition selection

Solution NMR Limitations:

  • Size constraints typically limiting NMR to proteins <30 kDa

  • Requirement for isotopic labeling (15N, 13C, 2H)

  • Spectral crowding in larger proteins making assignment difficult

  • Conformational dynamics complicating data interpretation

Cryo-EM Considerations:

  • Smaller proteins (<50 kDa) challenging to visualize without fusion partners

  • Sample heterogeneity affecting 3D reconstruction quality

  • Requirement for highly specialized equipment and expertise

  • Data processing complexity for novel protein structures

Strategic Approaches:

  • Utilize fragment-based approaches by identifying stable domains

  • Apply integrative structural biology combining multiple techniques

  • Consider membrane mimetics if the protein may associate with membranes

  • Use computational prediction to guide experimental design

  • Explore co-crystallization with binding partners or antibody fragments

For B. subtilis proteins specifically, leveraging the extensive genomic knowledge and available expression systems optimized for this organism can provide advantages in structural studies .

How can genetic knockouts and complementation studies help characterize uncharacterized B. subtilis proteins?

Genetic manipulation approaches provide critical functional insights for uncharacterized proteins:

Knockout Strategy Design:

  • Clean deletion vs. insertional inactivation considerations

  • Use of counter-selectable markers for scarless deletions

  • Construction of conditional mutants for essential genes

  • CRISPR-Cas9 approaches for precise genome editing

Phenotypic Analysis Framework:

  • Growth curve analysis under various conditions (temperature, pH, nutrients)

  • Stress resistance profiling (oxidative, osmotic, antibiotic)

  • Morphological characterization (microscopy, cell size, sporulation)

  • Metabolic phenotyping (carbon source utilization, metabolite profiling)

  • Global transcriptomic/proteomic changes in knockout strains

Complementation Approaches:

  • Integration vs. plasmid-based complementation considerations

  • Native promoter vs. inducible expression systems

  • Structure-function analysis through targeted mutations

  • Cross-species complementation to assess functional conservation

Synthetic Genetic Interactions:

  • Double knockout construction to identify genetic interactions

  • Synthetic lethality screening to identify functional pathways

  • Suppressor mutation analysis to identify compensatory mechanisms

B. subtilis is particularly amenable to these genetic approaches due to its natural competence and integration of DNA into its genome . Additionally, the availability of genome-minimized B. subtilis strains provides unique opportunities to study protein function in simplified genetic backgrounds .

How do I address protein insolubility issues when expressing B. subtilis uncharacterized proteins?

Insolubility is a common challenge when expressing uncharacterized proteins. A systematic troubleshooting approach includes:

Expression Condition Optimization:

  • Reduce expression temperature (16-25°C) to slow folding and prevent aggregation

  • Decrease inducer concentration to reduce expression rate

  • Use rich media supplements (e.g., casamino acids) to provide additional chaperone resources

  • Consider auto-induction media for gradual protein expression

Fusion Partner Strategies:

  • Test solubility-enhancing fusion partners (MBP, SUMO, TrxA, GST)

  • Compare N-terminal vs. C-terminal fusion configurations

  • Optimize linker length between fusion partner and target protein

  • Assess solubility before and after tag removal

Co-expression Approaches:

  • Co-express with chaperones (GroEL/GroES, DnaK/DnaJ/GrpE)

  • Co-express with potential binding partners if known

  • Implement dual plasmid systems with tunable expression levels

  • Consider specialized B. subtilis strains with enhanced chaperone expression

Buffer Optimization Matrix:

Buffer ComponentVariations to Test
pH5.0-9.0 in 0.5 increments
Salt (NaCl)0, 50, 100, 250, 500 mM
AdditivesGlycerol (5-20%), Arginine (50-500 mM), Proline (50-500 mM)
DetergentsTriton X-100 (0.01-0.1%), CHAPS (0.1-1%), DDM (0.01-0.1%)
Reducing agentsDTT (1-10 mM), TCEP (0.5-5 mM), β-ME (1-10 mM)

Refolding Strategies:
If inclusion bodies persist, implement on-column refolding or dilution refolding with optimized buffer conditions based on protein characteristics.

What strategies can overcome low expression yields of B. subtilis uncharacterized proteins?

Low expression yields of uncharacterized proteins can be addressed through:

Transcriptional Optimization:

  • Test different promoter systems (Pveg, PgroE, Pspac, PxylA) with varying strengths

  • Optimize ribosome binding site (RBS) sequence and spacing

  • Evaluate the effect of 5' untranslated region (UTR) sequences on expression

  • Consider genomic integration at different loci to assess position effects

Translational Enhancement:

  • Codon optimization based on B. subtilis preferred codons

  • Addition of translation enhancer sequences

  • Optimization of mRNA secondary structure in the translation initiation region

  • Evaluate different signal peptides if secretory expression is desired

Host Strain Selection:

  • Test expression in protease-deficient strains (e.g., WB800 derivatives)

  • Evaluate expression in genome-minimized strains that have shown superior performance

  • Consider auxotrophic strains with enhanced expression capabilities

  • Use strains with mutations in transcriptional regulators that might affect target gene expression

Scale-up Strategies:

  • Implement fed-batch fermentation to achieve higher cell densities

  • Optimize media composition based on cellular requirements

  • Consider using defined media for consistent expression results

  • Monitor dissolved oxygen levels and supplement as needed during high-density cultivation

Post-translational Stability Enhancement:

  • Add protease inhibitors during extraction and purification

  • Optimize cell lysis conditions to minimize proteolytic degradation

  • Consider lower temperature handling throughout the purification process

  • Evaluate buffer additives that enhance protein stability

How can I design effective functional assays for uncharacterized B. subtilis proteins with unknown activities?

Designing functional assays for proteins of unknown function requires a structured approach:

Bioinformatic-Guided Hypothesis Generation:

  • Sequence-based predictions of biochemical activity

  • Structural homology modeling to identify potential active sites

  • Analysis of conserved residues across homologs

  • Domain architecture comparison with characterized proteins

Activity Screening Approaches:

  • Generic enzyme class assays (hydrolase, transferase, oxidoreductase activities)

  • Substrate panels based on predicted biochemical function

  • Metabolite profiling of knockout vs. wild-type strains

  • Comparative activity assays with closely related characterized proteins

Protein-Protein Interaction Screening:

  • Pull-down assays coupled with mass spectrometry

  • Bacterial two-hybrid or yeast two-hybrid screening

  • Surface plasmon resonance with candidate interactors

  • Crosslinking coupled with mass spectrometry to capture transient interactions

In vivo Functional Assessment:

  • Phenotypic microarray analysis of knockout strains

  • Transcriptomic profiling to identify affected pathways

  • Fluorescent reporter assays for potential regulatory functions

  • Subcellular localization studies to inform function

Assay Development Workflow:

  • Generate hypotheses based on available data

  • Design preliminary assays to test broad activity classes

  • Refine assays based on initial results

  • Validate with positive and negative controls

  • Establish quantitative parameters (Km, Vmax, binding constants)

  • Confirm physiological relevance through in vivo studies

How can CRISPR-Cas9 technology be applied to study uncharacterized B. subtilis proteins?

CRISPR-Cas9 technology has revolutionized genetic manipulation in B. subtilis and offers powerful approaches for studying uncharacterized proteins:

Precise Genome Editing:

  • Scarless gene deletions to create clean knockouts

  • Introduction of point mutations to study structure-function relationships

  • Insertion of epitope tags for protein detection and purification

  • Creation of fluorescent protein fusions at genomic loci for localization studies

Regulatory Element Manipulation:

  • Promoter replacements to control expression levels

  • RBS modifications to alter translation efficiency

  • Terminator modifications to affect mRNA stability

  • UTR modifications to influence post-transcriptional regulation

High-Throughput Applications:

  • Multiplexed gene editing for pathway engineering

  • Creation of knockout libraries for phenotypic screening

  • Systematic domain deletion analysis

  • Scanning mutagenesis of coding regions

CRISPRi Applications:

  • Tunable gene repression through dCas9-based interference

  • Temporal control of gene expression

  • Study of essential genes where knockouts would be lethal

  • Simultaneous repression of multiple genes to study redundant functions

Implementation Strategy:

  • Design specific sgRNAs with minimal off-target effects

  • Optimize Cas9 expression for B. subtilis (codon optimization, appropriate promoters)

  • Develop efficient delivery methods (natural competence, electroporation)

  • Include appropriate selection markers and counter-selection systems

  • Verify edits through sequencing and phenotypic confirmation

This technology is particularly valuable for B. subtilis given its natural competence and recombination capabilities, allowing for efficient transformation and integration of editing templates .

What systems biology approaches can help decipher the function of uncharacterized B. subtilis proteins?

Systems biology provides holistic approaches to understand protein function within the cellular network:

Multi-omics Integration:

  • Transcriptomics to identify co-regulated genes

  • Proteomics to establish protein abundance and modifications

  • Metabolomics to detect changes in metabolic profiles

  • Fluxomics to measure metabolic flux alterations in knockout strains

Network Analysis Methods:

  • Protein-protein interaction network mapping

  • Genetic interaction networks through synthetic genetic arrays

  • Regulatory network inference from transcriptomic data

  • Metabolic network analysis to identify potential enzymatic roles

Computational Modeling Approaches:

  • Constraint-based metabolic models (e.g., Flux Balance Analysis)

  • Kinetic models of relevant pathways

  • Whole-cell models incorporating multiple cellular processes

  • Machine learning approaches to predict protein function from multi-omics data

Integration Framework:

Data TypeAnalysis ApproachFunctional Insights
TranscriptomicsDifferential expression, co-expression networksRegulatory relationships, functional pathways
ProteomicsAbundance changes, post-translational modificationsProtein activity, complex formation
MetabolomicsMetabolite profile changesEnzymatic activities, pathway involvement
PhenomicsGrowth, morphology, stress responsePhysiological role, environmental response
InteractomicsProtein-protein interactionsComplex membership, signaling pathways

Implementation Strategy:

  • Generate knockout or conditional mutant of the target gene

  • Perform multi-omics analyses under relevant conditions

  • Integrate datasets using computational tools

  • Identify significantly altered networks or pathways

  • Generate testable hypotheses about protein function

  • Validate predictions with targeted experiments

B. subtilis is particularly amenable to systems biology approaches due to its well-annotated genome and extensive existing datasets from previous studies .

How can synthetic biology approaches advance our understanding of uncharacterized B. subtilis proteins?

Synthetic biology offers innovative strategies to study uncharacterized proteins:

Minimal Genome Approaches:

  • Expression of the protein in genome-minimized B. subtilis strains

  • Systematic addition of genetic elements to determine minimal functional requirements

  • Creation of synthetic gene clusters to test hypothesized functions

  • B. subtilis has undergone successful genome minimization by 40%, providing valuable platforms for such studies

Protein Engineering Strategies:

  • Domain shuffling to create chimeric proteins with partial known functions

  • Directed evolution to enhance potential activities

  • Creation of synthetic protein scaffolds to test domain functions

  • Rational design based on structural predictions

Synthetic Circuit Design:

  • Creation of reporter systems linked to protein activity

  • Implementation of feedback loops to amplify phenotypic effects

  • Design of toggle switches to study dynamic protein functions

  • Construction of synthetic pathways to test metabolic roles

Orthogonal Expression Systems:

  • Non-native promoters and regulatory elements to control expression

  • Inducible systems for temporal control of expression

  • Orthogonal ribosomes for specialized translation

  • Alternative genetic codes to incorporate non-standard amino acids for functional studies

Implementation Framework:

  • Define the specific question about protein function

  • Design synthetic biological system to address the question

  • Model the system behavior computationally

  • Construct genetic parts and assemble into the designed system

  • Test system functionality through appropriate assays

  • Iterate design based on experimental outcomes

B. subtilis is an excellent chassis for synthetic biology approaches due to its genetic tractability, well-characterized expression systems, and capacity for high-level protein production .

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.