Recombinant Bacillus cereus UPF0213 protein BCE_0033 (BCE_0033)

Shipped with Ice Packs
In Stock

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 the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification 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 default glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
BCE_0033; UPF0213 protein BCE_0033
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-96
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacillus cereus (strain ATCC 10987 / NRS 248)
Target Names
BCE_0033
Target Protein Sequence
MEKNKHCFYV VECSDGSYYA GYTNHIEKRI ETHNSGKGAK YTRARLPVAL KYVEFHEDKR TAMQAEYYFK QLNRKQKEEY MQKGEPYVAT KKFSTK
Uniprot No.

Q&A

How should I design primers for cloning BCE_0033?

When designing primers for BCE_0033 cloning, begin by retrieving the complete gene sequence from genomic databases. Design primers that include appropriate restriction enzyme sites compatible with your expression vector of choice, typically incorporating a 5-6 nucleotide overhang before the restriction site to ensure efficient enzyme digestion. For example, in the cloning of EntD from B. cereus, researchers used primers with BamHI and EcoRI restriction sites with lowercase letters indicating the added restriction sites and uppercase letters matching the gene sequence (5′-ggatccAAATTCTAAAAATCTGTTGCTATAATG-3′ and 5′-gaattcTTCGCCCCCAGCTATTAGGACTA-3′) . Additionally, ensure primers have appropriate melting temperatures (typically 55-65°C) and verify they lack secondary structures that might impede PCR efficiency.

What purification strategy should I use for BCE_0033?

A multi-step purification strategy typically yields the best results. Begin with affinity chromatography using a suitable tag (His6, GST, or MBP) based on your expression construct. For BCE_0033, consider using a His-tag for initial capture, followed by ion exchange chromatography to remove remaining contaminants, and finally size exclusion chromatography to ensure homogeneity and remove aggregates. Throughout purification, monitor protein stability by testing different buffer conditions (varying pH 6.0-8.0, salt concentrations 100-500 mM NaCl, and adding glycerol 5-10% or reducing agents if necessary). Validate protein identity and purity using SDS-PAGE, Western blotting, and mass spectrometry techniques to confirm the correct molecular weight and sequence integrity, similar to the approaches used in identifying and characterizing other B. cereus proteins .

How can I verify the expression of BCE_0033 in recombinant systems?

Verification requires multiple complementary approaches. Start with SDS-PAGE analysis of cell lysates before and after induction to observe the appearance of a band at the expected molecular weight. Confirm protein identity through Western blotting using antibodies against the protein or any epitope tags. For definitive confirmation, perform mass spectrometry analysis to identify peptide fragments matching the expected BCE_0033 sequence. RT-PCR can verify transcriptional activity by measuring mRNA levels, as was done in EntD studies to confirm gene expression levels . Additionally, if available, activity assays specific to predicted functions of BCE_0033 can provide functional validation of proper protein expression.

What approaches can be used to determine BCE_0033 function through mutational studies?

To determine BCE_0033 function through mutational studies, first construct a BCE_0033 knockout mutant using homologous recombination techniques. Design a knockout construct that replaces the BCE_0033 gene with an antibiotic resistance marker, similar to the spectinomycin cassette approach used in EntD studies . Confirm gene disruption through PCR verification and RT-PCR to ensure the absence of transcripts. Then conduct comprehensive phenotypic characterization comparing the mutant to wild-type strains, examining growth kinetics across various media conditions, cell morphology through electron microscopy, metabolic profiles, stress responses, and virulence characteristics. Complementation studies using a plasmid-based expression system can verify that observed phenotypes are directly attributable to BCE_0033 deletion, though careful calibration of expression levels is critical as overexpression may not restore wild-type phenotypes .

How can proteomics be applied to understand BCE_0033's role in B. cereus physiology?

A comprehensive proteomics approach involves comparing the cellular and extracellular proteomes of wild-type and BCE_0033 knockout strains. Use label-free quantitative proteomics with liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) to identify proteins with differential abundance between strains. Sample cellular proteomes from multiple growth phases (early exponential, late exponential, and stationary phase) to capture temporal changes in protein expression profiles. Analyze data using statistical methods (p < 0.05) to identify significant changes in protein abundance levels. Categorize affected proteins by functional groups to identify biological processes influenced by BCE_0033, as was done with EntD where 308 and 79 proteins were identified as regulated in the cellular proteome and exoproteome, respectively . This approach can reveal cellular pathways and functions dependent on BCE_0033 activity.

What techniques should I use to investigate BCE_0033's potential role in B. cereus virulence?

Investigating BCE_0033's role in virulence requires a multi-faceted approach. Begin with cytotoxicity assays using relevant cell lines (such as Caco-2 cells for gastrointestinal toxicity) to compare wild-type and BCE_0033 mutant strains, measuring cell viability through MTT or LDH assays . Quantify expression levels of known virulence factors (Nhe, Hbl, CytK) using both proteomics and RT-PCR to assess BCE_0033's regulatory impact on toxin production. Examine bacterial adherence to epithelial cells and biofilm formation capacity using crystal violet staining and confocal microscopy. For in vivo relevance, consider infection models in appropriate organisms based on the specific disease association being investigated. Additionally, assess motility through swimming and swarming assays on semi-solid media, as altered motility can correlate with virulence potential in B. cereus, as observed with EntD disruption .

How can structural biology approaches be applied to understand BCE_0033 function?

A comprehensive structural biology investigation begins with computational prediction of protein structure using homology modeling or ab initio methods to generate initial structural hypotheses. For experimental validation, express and purify BCE_0033 in a form suitable for structural studies (typically requiring >95% purity and high concentration). Apply X-ray crystallography to obtain high-resolution structures by screening crystallization conditions systematically. Alternatively, use nuclear magnetic resonance (NMR) spectroscopy for proteins under 25 kDa or cryo-electron microscopy (cryo-EM) for larger complexes. Complement these approaches with small-angle X-ray scattering (SAXS) to examine protein conformation in solution. Identify potential binding partners through pull-down assays coupled with mass spectrometry or yeast two-hybrid screening. Structure-function relationships can be further explored by site-directed mutagenesis of predicted functional residues, followed by activity assays to correlate structural features with biochemical function.

What methods can determine if BCE_0033 interacts with cell wall components?

To investigate potential interactions between BCE_0033 and cell wall components, employ a combination of in vitro and in vivo approaches. Begin with co-sedimentation assays using purified BCE_0033 and isolated cell wall fragments, analyzing bound fractions by SDS-PAGE and Western blotting. For higher resolution analysis, conduct surface plasmon resonance (SPR) to measure binding kinetics with purified peptidoglycan components. In cellular contexts, use fluorescently tagged BCE_0033 to visualize localization patterns through confocal microscopy, paying particular attention to cell envelope association. Electron microscopy with immunogold labeling can provide nanometer-scale resolution of protein localization. Analyze effects of BCE_0033 deletion on cell wall ultrastructure using transmission electron microscopy, looking for alterations in cell wall thickness or organization similar to those observed in studies of other B. cereus proteins . Additionally, assess changes in autolytic rate and sensitivity to cell wall-targeting antibiotics, which can indicate functional relationships with cell wall metabolism.

How should I analyze the transcriptional regulation of BCE_0033?

Analyzing BCE_0033 transcriptional regulation requires characterizing its promoter region and regulatory elements. Begin by identifying the transcriptional start site using 5' RACE (Rapid Amplification of cDNA Ends) to precisely map the initiation point, as was done with EntD where a transcriptional start site was identified 26 bp upstream of the translational start codon . Analyze the upstream region for putative promoter elements and regulatory protein binding sites using bioinformatic tools. Construct transcriptional reporter fusions (using fluorescent proteins or luciferase) to measure promoter activity under various environmental conditions, including different growth phases, temperatures, pH levels, and nutrient availability. Employ chromatin immunoprecipitation (ChIP) followed by sequencing to identify regulatory proteins that physically interact with the BCE_0033 promoter region in vivo. RT-qPCR can quantify BCE_0033 expression levels across different conditions to establish an expression profile, which may reveal functional clues based on co-regulation patterns with genes of known function.

What bioinformatic approaches can predict BCE_0033 function?

A comprehensive bioinformatic analysis begins with sequence-based predictions using tools like BLAST, HMMER, and InterProScan to identify conserved domains and sequence motifs that might suggest function. For BCE_0033, as a member of the UPF0213 family, look for structural domains similar to those found in related proteins, such as the SH3_3 domains and cell wall binding domains identified in EntD . Apply structural prediction algorithms (AlphaFold, I-TASSER) to generate three-dimensional models that can reveal functional sites not apparent from sequence alone. Conduct genomic context analysis to identify conserved gene neighborhoods and operonic structures, as genes with related functions often cluster together. Metabolic pathway mapping can situate BCE_0033 within the cellular biochemical network. Additionally, perform phylogenetic analysis to trace the evolutionary history of BCE_0033, identifying orthologs in related species that might have characterized functions. Gene co-expression network analysis using publicly available transcriptomic datasets can reveal functional associations based on expression patterns.

How can I effectively characterize protein-protein interactions involving BCE_0033?

Characterizing BCE_0033's protein-protein interactions requires multiple complementary approaches. Begin with affinity purification coupled to mass spectrometry (AP-MS) by expressing tagged BCE_0033 in B. cereus, followed by pull-down experiments to identify co-purifying proteins. Validate key interactions using targeted methods such as co-immunoprecipitation with specific antibodies. For direct binary interactions, employ yeast two-hybrid or bacterial two-hybrid systems, which can detect interactions in a cellular context. In vitro validation can be performed using surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) to determine binding affinities and thermodynamic parameters. For spatial context, use proximity labeling approaches like BioID or APEX2, where BCE_0033 is fused to an enzyme that biotinylates nearby proteins, allowing identification of the proximal proteome. Map interaction domains through truncation or point mutation analyses to identify specific regions critical for protein-protein binding. Functional significance can be assessed by disrupting key interactions through targeted mutations and observing phenotypic effects.

What approaches can elucidate BCE_0033's role in metabolic pathways?

To elucidate BCE_0033's role in metabolic pathways, implement a systems biology approach combining multiple techniques. Compare metabolomic profiles of wild-type and BCE_0033 knockout strains using liquid or gas chromatography coupled to mass spectrometry (LC-MS or GC-MS) to identify metabolites with altered abundance. Measure key metabolic parameters including growth rate, substrate consumption, and byproduct formation across different carbon sources and growth conditions. Stable isotope labeling experiments can trace carbon flux through central metabolic pathways, identifying specific reactions affected by BCE_0033 deletion. Integrate these findings with transcriptomic and proteomic data to construct a comprehensive metabolic network model. As observed in the EntD study, disruption of B. cereus proteins can significantly impact central metabolism, including glycolysis and TCA cycle activity, which was evident from changes in enzyme abundance and metabolic outputs such as acetate production . Enzyme activity assays targeting pathways identified through omics approaches can provide functional validation of metabolic roles.

How do I design experiments to investigate BCE_0033's potential role in stress response?

Design a comprehensive stress response investigation by first subjecting wild-type and BCE_0033 knockout strains to a range of stressors, including oxidative stress (H₂O₂, paraquat), heat shock, cold shock, osmotic stress (NaCl, sorbitol), acid stress, and antimicrobial compounds. Measure survival rates, growth kinetics, and recovery times under each condition. For oxidative stress specifically, quantify intracellular reactive oxygen species (ROS) levels using fluorescent probes and measure activities of antioxidant enzymes such as catalase, superoxide dismutase, and peroxidase. Proteomic analysis comparing stress-exposed wild-type and mutant strains can identify stress response proteins regulated by BCE_0033, similar to how EntD was found to affect numerous proteins involved in cellular stress responses . Employ transcriptomics to identify genes differentially expressed during stress exposure, focusing on known stress response regulons. Monitor morphological changes under stress conditions using microscopy techniques. Additionally, assess the formation of stress-induced structures such as endospores, as sporulation efficiency can be an important measure of stress adaptation in Bacillus species.

How can I address poor expression of recombinant BCE_0033?

Poor expression of BCE_0033 may stem from multiple factors requiring systematic troubleshooting. First, review codon optimization for the expression host, as rare codons can significantly impede translation efficiency. Consider using specialized strains like Rosetta for E. coli expression systems or codon-optimize the gene sequence. Test alternative expression vectors with different promoters, as some proteins express better under T7, tac, or arabinose-inducible systems. Modify culture conditions by reducing induction temperature (16-25°C), decreasing inducer concentration, and extending expression time to promote proper folding. Expression as a fusion protein with solubility-enhancing partners (MBP, SUMO, or Trx) can improve yield. For potentially toxic proteins, implement tightly controlled expression systems or consider cell-free protein synthesis. If DNA sequencing reveals no errors but expression remains poor, examine mRNA levels through RT-PCR to determine if the issue lies at the transcriptional or translational level, a diagnostic approach used in studies of other B. cereus proteins .

What strategies can I use when BCE_0033 knockout creation is unsuccessful?

When BCE_0033 knockout creation proves challenging, first consider whether BCE_0033 might be essential for viability, which would explain the inability to generate null mutants. To test this hypothesis, attempt to create conditional mutants using inducible promoters or degradation tags that allow controlled depletion rather than complete elimination. Alternative approaches include creating partial deletions or point mutations that diminish function without completely removing the gene. CRISPR interference (CRISPRi) can repress gene expression without modifying the genome and may be more successful for essential genes. If technical issues are suspected, optimize transformation efficiency by testing different competent cell preparation methods and electroporation parameters. Verify homologous recombination regions for secondary structures or repetitive elements that might impede crossover events. Consider using different antibiotic resistance markers if the current selection system shows poor efficiency. The challenges encountered in complementation of the EntD mutant, where plasmid-based expression led to overexpression that failed to restore wild-type phenotypes , highlight the importance of expression level control in genetic manipulation experiments.

How should I interpret contradictory results between in vitro and in vivo BCE_0033 studies?

Contradictory results between in vitro and in vivo BCE_0033 studies require careful analysis and reconciliation. Begin by critically examining methodological differences that might explain the discrepancies, including protein concentration, buffer conditions, presence of cofactors, and post-translational modifications that may be present in vivo but absent in vitro. Consider the complexity of cellular environments where BCE_0033 may interact with multiple partners simultaneously, while in vitro studies typically isolate specific interactions. Time-resolved studies can reveal whether differences result from kinetic effects that manifest differently in the more dynamic cellular environment. Validate protein folding and activity in vitro to ensure the recombinant protein accurately represents the native form. If studying regulatory functions, remember that regulatory networks may compensate for BCE_0033 absence in vivo through alternative pathways, masking effects that are apparent in simplified in vitro systems. This phenomenon was observed with EntD, where the deletion of entD led to increased abundance of related proteins (EntA and EntC), potentially offsetting some effects of the deletion . Ultimately, both approaches provide valuable complementary insights, with in vitro studies offering mechanistic detail and in vivo work providing physiological context.

What considerations are important when scaling up BCE_0033 production for structural studies?

Scaling up BCE_0033 production for structural studies requires optimization at every stage of the process. Begin with expression system refinement, potentially moving from shake flasks to bioreactors with controlled dissolved oxygen, pH, and feeding strategies. For B. cereus proteins, high cell density cultivation using fed-batch processes can significantly increase biomass and subsequent protein yield. Optimize cell lysis methods, comparing sonication, high-pressure homogenization, and chemical lysis to identify the approach that maximizes recovery while preserving protein structure. Implement a rational purification pipeline with high-capacity chromatography media and consider automated systems for reproducibility. Throughout scaling, monitor protein quality using dynamic light scattering to assess monodispersity and circular dichroism to verify secondary structure integrity. For crystallography purposes, identify stabilizing buffer conditions through thermal shift assays and storage conditions that maintain long-term stability. Concentrate protein carefully, monitoring for aggregation through size exclusion chromatography. If protein yield remains insufficient, consider expression in eukaryotic systems like insect cells or yeast, which can provide higher yields for certain proteins while maintaining proper folding.

How can I differentiate between direct and indirect effects of BCE_0033 on cellular processes?

Differentiating between direct and indirect effects requires multiple lines of evidence and controlled experimental designs. Direct physical interactions can be demonstrated through techniques like surface plasmon resonance (SPR), isothermal titration calorimetry (ITC), or pull-down assays using purified components. For enzymatic activities, in vitro reconstitution with purified components can establish direct effects. Time-course studies can help establish causality, as direct effects typically occur more rapidly than indirect ones. Dosage dependency analysis, where cellular responses are measured across a range of BCE_0033 concentrations or expression levels, can help distinguish direct from indirect effects. Point mutations in BCE_0033 that specifically disrupt particular functions while preserving others can isolate direct effects on specific pathways. For regulatory effects, chromatin immunoprecipitation (ChIP) can identify direct DNA binding. Similarly, RNA immunoprecipitation can establish direct RNA interactions if BCE_0033 functions post-transcriptionally. In studies of other B. cereus proteins like EntD, researchers distinguished direct from indirect effects by correlating proteomic changes with specific phenotypic alterations and validating key findings with targeted experiments . Computational network analysis can also help predict the likely direct targets of BCE_0033 based on interaction patterns.

What are promising approaches for identifying BCE_0033 substrates or binding partners?

Identifying BCE_0033 substrates or binding partners requires a multi-technique approach beginning with unbiased screening methods. Chemical cross-linking coupled with mass spectrometry (XL-MS) can capture transient interactions by covalently linking BCE_0033 to its partners in vivo before analysis. Proximity-dependent biotin identification (BioID) or APEX2 proximity labeling can map the protein neighborhood around BCE_0033 in living cells. For substrate identification, develop activity-based protein profiling probes if BCE_0033 is suspected to have enzymatic activity. Phage display or synthetic peptide libraries can identify specific binding motifs recognized by BCE_0033. Once candidate partners are identified, validate interactions using fluorescence resonance energy transfer (FRET), bioluminescence resonance energy transfer (BRET), or split-protein complementation assays in vivo. Structural studies of BCE_0033 in complex with binding partners using X-ray crystallography or cryo-EM can provide atomic-level details of interaction interfaces. Computational prediction methods, including protein-protein docking and molecular dynamics simulations, can guide experimental designs by suggesting likely interaction modes based on structural complementarity.

How can systems biology approaches enhance our understanding of BCE_0033 function?

Systems biology approaches can provide a holistic understanding of BCE_0033 function by integrating multiple layers of biological information. Implement multi-omics studies combining transcriptomics, proteomics, metabolomics, and potentially lipidomics to create comprehensive network models that position BCE_0033 within cellular pathways. Time-resolved analyses across different growth phases and environmental conditions can reveal dynamic relationships, similar to how EntD's impact was found to vary across growth phases . Apply network inference algorithms to identify regulatory relationships and functional modules associated with BCE_0033 activity. Flux balance analysis can model metabolic consequences of BCE_0033 perturbation and generate testable hypotheses about its metabolic roles. Leverage comparative systems biology by studying BCE_0033 homologs across different Bacillus species to identify conserved and species-specific functions. Machine learning approaches can identify patterns in large-scale datasets that might not be apparent through conventional analysis. Genome-scale genetic interaction mapping through technologies like CRISPRi can identify genes that exhibit synthetic lethality or suppression relationships with BCE_0033, providing functional context and revealing pathway connections.

What considerations are important when developing BCE_0033 as a potential therapeutic target?

Developing BCE_0033 as a therapeutic target requires systematic evaluation of several key factors. First, establish target validation by demonstrating that BCE_0033 inhibition leads to desired antimicrobial effects with minimal off-target consequences. Assess essentiality across diverse B. cereus strains to ensure broad-spectrum efficacy and evaluate conservation in other bacterial species to assess selectivity potential. Resolve the three-dimensional structure of BCE_0033 to identify druggable pockets suitable for small molecule binding. Develop high-throughput screening assays to test compound libraries, ideally based on a well-defined biochemical activity of BCE_0033. Evaluate resistance development potential through in vitro evolution experiments and whole genome sequencing of spontaneous resistant mutants. Consider combination therapy approaches if BCE_0033 inhibition alone provides insufficient antimicrobial activity. For in vivo efficacy, establish appropriate animal infection models that recapitulate human B. cereus infections. Pharmacokinetic and pharmacodynamic studies should assess compound distribution to infection sites. Since B. cereus proteins like EntD can influence virulence factor production , targeting BCE_0033 might provide an anti-virulence strategy rather than directly killing bacteria, potentially reducing selection pressure for resistance development.

How might BCE_0033 function relate to other uncharacterized proteins in B. cereus?

BCE_0033's function may interconnect with other uncharacterized proteins through shared pathways, protein complexes, or regulatory networks. To explore these relationships, implement co-expression analysis across diverse conditions to identify genes with similar expression patterns, suggesting functional relationships. Construct protein-protein interaction networks through systematic screening approaches to identify physical interactions between BCE_0033 and other uncharacterized proteins. Perform comparative genomic analysis to identify conserved genomic neighborhoods and potential operonic structures that might include multiple uncharacterized genes functioning in related processes. Create multiple gene knockout strains to identify synthetic phenotypes that emerge only when BCE_0033 is deleted in combination with other genes. Proteomic analysis of BCE_0033 deletion strains can reveal other uncharacterized proteins whose abundance changes significantly, suggesting functional connections . Apply hierarchical clustering to large-scale phenotypic data to identify uncharacterized proteins that produce similar phenotypic signatures when disrupted. These approaches can help define functional modules containing BCE_0033 and other uncharacterized proteins, potentially assigning them to shared biological processes even before their precise molecular functions are determined.

What emerging technologies could advance BCE_0033 research in the next five years?

Emerging technologies poised to advance BCE_0033 research include several cutting-edge approaches. Cryo-electron tomography can visualize BCE_0033 in its native cellular context at near-atomic resolution, revealing spatial organization and protein complexes without isolation or crystallization. AlphaFold2 and other AI-powered structure prediction tools will continue improving, potentially enabling accurate modeling of BCE_0033 complexes with interaction partners. Single-cell proteomics technologies can reveal cell-to-cell variability in BCE_0033 expression and function within bacterial populations. CRISPR-based technologies beyond gene editing, such as CRISPRi for transcriptional repression and CRISPRa for activation, offer precise control over BCE_0033 expression. Microfluidic systems coupled with time-lapse microscopy can track BCE_0033 dynamics in individual cells across changing environments. Nanopore direct RNA sequencing may reveal post-transcriptional regulation affecting BCE_0033. Advanced mass spectrometry techniques like top-down proteomics and hydrogen-deuterium exchange mass spectrometry (HDX-MS) can provide detailed information about BCE_0033 proteoforms and dynamic structural changes. Synthetic biology approaches, including minimal cell systems, could reveal the essentiality of BCE_0033 in defined genetic backgrounds, while genome-wide CRISPRi screens could systematically map genetic interactions across the entire B. cereus genome.

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.