Recombinant Bacillus cereus UPF0145 protein BCE_5284 (BCE_5284)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
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. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on 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. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us for preferential development.
Synonyms
BCE_5284; UPF0145 protein BCE_5284
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-103
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacillus cereus (strain ATCC 10987 / NRS 248)
Target Names
BCE_5284
Target Protein Sequence
MIVTTTSTIQ GKEIIEYIDI VNGEAIMGAN IVRDLFASVR DVVGGRAGAY ESKLKEARDI AMEEMKTFAR QKNANAIVGI DVDYEVVREG MLMVAVSGTA VRI
Uniprot No.

Q&A

What is Bacillus cereus and what are its major virulence factors?

Bacillus cereus is a gram-positive, spore-forming bacterium widely distributed in various environments and food products. It causes food poisoning with diverse symptomatology through several key virulence factors:

  • Hemolysin BL (HBL): A tripartite toxin composed of B, L1, and L2 components encoded by the genes hblA, hblD, and hblC, respectively

  • Non-hemolytic enterotoxin (NHE): A three-component toxin encoded by nheA, nheB, and nheC genes

  • Cytotoxin K: A single-component toxin encoded by the cytK gene

  • Emetic toxin: Encoded by the cesB gene

  • Enterotoxin FM: Encoded by the entFM gene

The prevalence of these toxin genes varies significantly among B. cereus strains, with entFM present in 100% of isolates while the cesB gene appears in only 7% of isolates .

How does the molecular architecture of B. cereus toxins contribute to their function?

The molecular architecture of B. cereus toxins, particularly hemolysin BL (HBL), is critical to their function. HBL operates through a sequential assembly mechanism:

  • The three subunits (B, L1, and L2) bind to the cell membrane in a precise linear order

  • This ordered assembly is essential for forming a functional lytic pore

  • The resulting pore disrupts membrane integrity, causing potassium efflux

  • This potassium efflux triggers activation of the NLRP3 inflammasome

  • Activated NLRP3 inflammasome induces secretion of pro-inflammatory cytokines IL-1β and IL-18

  • The process culminates in pyroptosis, an inflammatory form of cell death

This multi-step mechanism demonstrates how protein architecture directly influences cellular outcomes and disease pathogenesis.

What genetic diversity exists among B. cereus strains regarding toxin production?

B. cereus strains exhibit remarkable genetic diversity in their toxin production capabilities:

Table 1: Distribution of Virulence Genes in B. cereus Food Isolates

Virulence GenePrevalence (%)
entFM100
nheB99
nheC94
nheA89
cytK68
hblD50
hblC49
hblA46
cesB7

Analysis of 368 B. cereus isolates identified 38 distinct virulence gene profiles. The most prevalent profile (33% of strains) contained eight virulence genes: hblA-hblC-hblD-nheA-nheB-nheC-entFM-cytK. Only two isolates harbored all nine virulence genes tested, while 27 isolates contained just three virulence genes each .

Multilocus sequence typing (MLST) further revealed that these 368 isolates belonged to 192 different sequence types (STs), including 93 newly identified STs, with ST26 being the most common .

How prevalent is B. cereus contamination in food products?

The prevalence of B. cereus in food products presents a significant public health concern:

Table 2: B. cereus Contamination in Ready-to-Eat Foods

Quantitative analysis revealed that while most contaminated samples (68%) contained moderate levels (3-1100 MPN/g) of B. cereus, 10% of positive samples exceeded 1100 MPN/g . This is particularly concerning for ready-to-eat foods that are consumed without further heat treatment.

What experimental approaches are used for initial characterization of novel B. cereus proteins?

Initial characterization of novel B. cereus proteins typically involves:

  • Gene identification and sequence analysis: Utilizing genomic databases to identify the gene of interest and analyze its sequence conservation across bacterial strains

  • Expression profiling: Determining expression conditions through RT-PCR or RNA-seq analysis

  • Protein expression and purification: Cloning the gene into expression vectors, producing recombinant protein in bacterial systems, and purifying using affinity chromatography

  • Structural characterization: Employing circular dichroism, X-ray crystallography, or NMR spectroscopy

  • Functional screening: Testing for enzymatic activity, protein-protein interactions, or cellular effects

  • Localization studies: Determining cellular localization using fluorescent protein tags or immunofluorescence microscopy

These approaches provide fundamental insights into protein function before advancing to more specialized investigations .

How do B. cereus toxins activate the NLRP3 inflammasome at the molecular level?

B. cereus toxins, particularly HBL, activate the NLRP3 inflammasome through a mechanistically precise pathway:

  • The three components of HBL (B, L1, and L2) assemble sequentially on the cell membrane

  • The assembled complex forms a lytic pore that disrupts membrane integrity

  • This pore formation induces potassium (K+) efflux from the cell

  • The K+ efflux serves as the primary trigger for NLRP3 inflammasome activation

  • Activated NLRP3 recruits the adaptor protein ASC, forming visible "specks" - a hallmark of inflammasome assembly

  • The ASC specks recruit pro-caspase-1, leading to its activation

  • Active caspase-1 processes pro-IL-1β and pro-IL-18 into their mature forms

  • Caspase-1 also cleaves gasdermin D, releasing its N-terminal fragment

  • The gasdermin D N-terminal domain forms pores in the plasma membrane

  • These pores allow release of mature IL-1β and IL-18 and induce pyroptotic cell death

This mechanism is specific to NLRP3, as demonstrated by impaired inflammasome activation in NLRP3-deficient BMDMs and the inhibitory effect of the NLRP3-specific inhibitor MCC950. Importantly, the production of inflammasome-independent cytokines (TNF and KC/CXCL1) and the phosphorylation of IκB and ERK remain unaffected in the absence of NLRP3, confirming pathway specificity .

What methodological approaches provide optimal results for studying B. cereus protein interactions?

Studying B. cereus protein interactions requires multiple complementary methodologies:

  • In vitro binding assays:

    • Pull-down assays using purified recombinant proteins

    • Surface plasmon resonance (SPR) for kinetic measurements

    • Isothermal titration calorimetry (ITC) for thermodynamic parameters

  • Structural biology approaches:

    • X-ray crystallography of protein complexes

    • Cryo-electron microscopy for larger assemblies

    • NMR spectroscopy for dynamic interactions

  • Cellular interaction studies:

    • Fluorescence resonance energy transfer (FRET)

    • Bimolecular fluorescence complementation (BiFC)

    • Proximity ligation assay (PLA)

  • Cross-linking mass spectrometry:

    • Identification of protein-protein interaction interfaces

    • Detection of transient interactions

  • Functional validation:

    • Mutagenesis of putative interaction sites

    • Competitive inhibition studies

    • Cell-based functional assays measuring toxin activity

For toxins like HBL, assembly studies measuring sequential binding to cell membranes provide critical insights into the mechanism of pore formation and subsequent cellular effects .

How does pyroptosis contribute to B. cereus pathogenesis?

Pyroptosis plays a multifaceted role in B. cereus pathogenesis:

  • Cellular characteristics: B. cereus toxins induce distinctive pyroptotic features including:

    • Loss of membrane integrity

    • Cytoplasm rounding

    • Centralized nucleus in a deflated cell body

    • Loss of cytoplasmic content

    • Nuclear condensation

  • Bacterial dissemination: During pyroptosis, phagocytosed bacteria are expelled following membrane rupture, potentially facilitating bacterial spread .

  • Inflammatory amplification: Pyroptosis releases cellular contents including DAMPs (damage-associated molecular patterns), amplifying inflammation beyond the initial infected cells.

  • Host survival impact: Excessive activation of the HBL-responsive NLRP3 inflammasome drives rapid host mortality, suggesting that while inflammation is protective, hyperactivation becomes detrimental .

  • Therapeutic implications: Pharmacological inhibition of NLRP3 using MCC950 prevents B. cereus-induced lethality, demonstrating that modulating the inflammatory response rather than targeting the bacteria directly can be an effective strategy .

These findings illustrate that pyroptosis represents a double-edged sword in host defense, necessary for pathogen control but potentially fatal when excessive.

What are the challenges in distinguishing functional roles between similar B. cereus proteins?

Researchers face several challenges when distinguishing functional roles between similar B. cereus proteins:

  • Genetic redundancy: B. cereus contains multiple toxin systems (HBL, NHE, Cytotoxin K) with potentially overlapping functions. For example, while HBL was identified as the primary inflammasome activator, isolates lacking HBL but expressing NHE or Cytotoxin K may still exhibit virulence through alternative mechanisms .

  • Strain variation: The 38 different virulence gene profiles identified across B. cereus isolates create a complex experimental landscape for determining protein-specific effects .

  • Multi-component systems: Toxins like HBL require all three components for full activity, complicating studies that may inadvertently use incomplete component sets .

  • Technical limitations:

    • Purification difficulties for membrane-active proteins

    • Cross-reactivity of antibodies between similar protein families

    • Challenges in creating clean genetic knockouts without polar effects

  • Methodological solutions:

    • Creating isogenic mutants lacking specific genes (e.g., ΔHbl B. cereus)

    • Using neutralizing antibodies against specific components

    • Employing heterologous expression systems with defined components

    • Utilizing multi-omics approaches to capture system-wide effects

These approaches helped researchers definitively identify HBL as the key inflammasome-activating factor, distinguishing it from NHE and Cytotoxin K despite their similar properties .

How can antimicrobial resistance patterns inform therapeutic approaches for B. cereus infections?

Antimicrobial resistance (AMR) patterns in B. cereus provide critical insights for therapeutic development:

Table 3: Antimicrobial Resistance Rates in B. cereus Isolates

AntibioticResistance Rate (%)
Penicillin (P)100.00
Ampicillin (AMP)99.73
Amoxicillin-clavulanic acid (AMC)97.83
Rifampin (RD)93.21
Cefoxitin (FOX)95.38
Cephalothin (KF)82.34
Cefotetan (CTT)13.59
Quinupristin-dalfopristin (QD)19.57

These resistance patterns suggest:

  • β-lactam ineffectiveness: Near-universal resistance to β-lactams indicates these antibiotics should be avoided for B. cereus infections.

  • Alternative therapeutic targets: The high resistance rates to conventional antibiotics highlight the need for alternative approaches, such as:

    • Targeting toxin production through quorum sensing inhibitors

    • Neutralizing toxin activity with antibodies or decoy receptors

    • Modulating host response with inflammasome inhibitors like MCC950

  • Host-directed therapy potential: The demonstration that MCC950 (NLRP3 inhibitor) prevents B. cereus-induced lethality suggests that targeting host inflammatory pathways may be more effective than attempting to overcome antimicrobial resistance .

  • Surveillance importance: The genetic diversity of B. cereus (192 different sequence types) emphasizes the need for ongoing antimicrobial resistance surveillance to detect emerging resistance patterns .

What are the optimal protocols for detecting and quantifying B. cereus in experimental samples?

Detection and quantification of B. cereus requires systematic protocols:

  • Qualitative detection protocol:

    • Homogenize 25g sample in 225 mL Trypticase Soy Broth with polymyxin

    • Incubate 48±2 hours at 30±2°C

    • Streak resulting cultures onto mannitol egg yolk polymyxin agar plates (MYP)

    • Incubate MYP plates 24 hours at 30°C

    • Transfer presumptive colonies to chromogenic B. cereus agar plates

    • Confirm identification through biochemical characterization

  • Quantitative detection using Most Probable Number (MPN) method:

    • Prepare sample dilutions (10⁻¹, 10⁻², 10⁻³)

    • Inoculate three tubes per dilution with 1 mL each

    • Incubate 48±2 hours at 30±2°C

    • Observe for turbidity characteristic of B. cereus growth

    • Confirm positive tubes by streaking onto MYP agar

    • Calculate MPN using standard MPN tables

  • Molecular detection methods:

    • Extract DNA using specialized kits for gram-positive bacteria

    • Perform PCR targeting species-specific markers (16S rRNA, groEL)

    • Use multiplex PCR for simultaneous detection of virulence genes

    • Employ real-time PCR for quantification

    • Sequence amplicons for confirmation when necessary

These protocols follow standards established by the U.S. Food and Drug Administration and the National Food Safety Standard of China.

How should researchers approach genetic characterization of B. cereus strains?

Comprehensive genetic characterization of B. cereus strains involves multiple techniques:

  • Virulence gene profiling:

    • Extract genomic DNA using kits optimized for gram-positive bacteria

    • Perform PCR detection of key virulence genes:

      • HBL components: hblA, hblC, hblD

      • NHE components: nheA, nheB, nheC

      • Single-component toxins: cytK, entFM

      • Emetic toxin: cesB

    • Analyze PCR products through gel electrophoresis

  • Strain differentiation:

    • ERIC-PCR (Enterobacterial Repetitive Intergenic Consensus PCR) fingerprinting

    • Analysis of fingerprints using specialized software like Bionumerics

    • Critical for excluding clonal isolates from the same sample

  • Evolutionary relationship analysis:

    • Multilocus Sequence Typing (MLST) targeting seven housekeeping genes:

      • glp, gmk, ilv, pta, pur, pyc, and tpi

    • Sequence analysis and allele number assignment via PubMLST database

    • Determination of Sequence Types (STs) based on allele combinations

    • Identification of Clonal Complexes (CCs) using geoBURST analysis

    • Construction of minimum spanning trees to visualize evolutionary relationships

  • Whole genome sequencing:

    • Provides comprehensive genetic information

    • Enables identification of novel virulence factors

    • Allows core genome MLST for higher resolution typing

    • Facilitates comparative genomics across strains

These approaches provide complementary information about virulence potential, genetic diversity, and evolutionary relationships of B. cereus strains.

What cell culture models are most appropriate for studying B. cereus protein functions?

Selection of appropriate cell culture models is critical for B. cereus protein research:

  • Immune cell models:

    • Bone marrow-derived macrophages (BMDMs): Primary model for studying inflammasome activation, with both wild-type and NLRP3-deficient (Nlrp3-/-) variants available

    • THP-1 human monocytic cells: Useful for human-specific responses

    • Dendritic cells: For examining antigen presentation and adaptive immune activation

  • Intestinal epithelial models:

    • Caco-2 cells: Model intestinal barrier and toxin translocation

    • HT-29 cells: Alternative intestinal epithelial model

    • Intestinal organoids: Advanced 3D model incorporating multiple cell types

  • Specialized experimental systems:

    • Transwell systems: For studying barrier function and toxin translocation

    • Co-culture systems: Combining epithelial and immune cells

    • Ex vivo tissue explants: For more physiologically relevant responses

  • Experimental parameters:

    • For studying purified toxins: Control protein concentration (typically 0.1-10 μg/mL)

    • For bacterial infection: Optimize multiplicity of infection (MOI)

    • Include appropriate controls:

      • Heat-inactivated toxins

      • Isogenic bacterial mutants (e.g., ΔHbl B. cereus)

      • Specific inhibitors (e.g., MCC950 for NLRP3)

  • Readout assays:

    • Western blot analysis for protein processing

    • ELISA for cytokine quantification

    • Microscopy for morphological changes

    • Cell viability assays (MTT, LDH release)

    • Real-time monitoring using systems like IncuCyte

Selection of the appropriate model depends on the specific research question, with consideration of species-specific differences in immune responses.

What are the methodological considerations for measuring inflammasome activation by B. cereus toxins?

Comprehensive assessment of inflammasome activation by B. cereus toxins requires multiple methodological approaches:

  • Protein processing analysis:

    • Western blot detection of:

      • Caspase-1 processing (p20 and p10 fragments)

      • IL-1β processing (mature p17 form)

      • Gasdermin D cleavage to N-terminal pore-forming domain

    • Sample preparation must preserve fragile processed forms

  • Cytokine quantification:

    • ELISA measurement of secreted IL-1β and IL-18

    • Include inflammasome-independent cytokines (TNF, KC/CXCL1) as controls

    • Consider multiplex cytokine analysis for broader inflammatory profile

  • ASC speck formation:

    • Immunofluorescence microscopy to visualize ASC specks

    • Flow cytometry for quantitative analysis

    • Time-course studies to capture optimal speck formation

  • Cell death assessment:

    • LDH release assay for membrane permeabilization

    • Propidium iodide uptake for cell death quantification

    • Morphological characterization by microscopy:

      • Light microscopy for general features

      • Transmission electron microscopy for ultrastructural changes

    • Real-time monitoring using IncuCyte system

  • Genetic validation:

    • Comparison between wild-type and inflammasome-deficient cells

    • siRNA knockdown or CRISPR-Cas9 targeting of specific components

    • Reconstitution experiments in deficient cells

  • Pharmacological confirmation:

    • NLRP3 inhibitor MCC950

    • Caspase-1 inhibitors

    • K+ efflux inhibition by high extracellular K+

    • ROS inhibitors to test oxidative stress contribution

Combining these approaches provides robust validation of inflammasome activation mechanisms and helps distinguish between different inflammasome types.

What purification strategies yield optimal results for B. cereus proteins?

Purification of B. cereus proteins requires tailored strategies based on protein properties:

  • Expression system selection:

    • E. coli: Most common for non-toxic proteins, using strains like BL21(DE3)

    • B. subtilis: For proteins requiring gram-positive cellular machinery

    • Insect cells: For complex proteins requiring eukaryotic processing

    • Cell-free systems: For highly toxic proteins

  • Optimization parameters:

    • Induction conditions (temperature, inducer concentration, duration)

    • Codon optimization for expression host

    • Fusion tags selection (His, GST, MBP, SUMO)

    • Solubility enhancement through chaperone co-expression

  • Purification workflow for secreted toxins:

    • Affinity chromatography as initial capture step

    • Ion exchange chromatography for intermediate purification

    • Size exclusion chromatography as polishing step

    • Endotoxin removal for proteins used in cellular assays

  • Special considerations for multi-component toxins (like HBL):

    • Individual purification of each component

    • Validation of purity using SDS-PAGE and western blotting

    • Functional testing of individual components and reconstituted complexes

    • Analytical ultracentrifugation to verify complex formation

  • Quality control measures:

    • Mass spectrometry for identity confirmation

    • Circular dichroism for secondary structure verification

    • Dynamic light scattering for homogeneity assessment

    • Limulus amebocyte lysate (LAL) assay for endotoxin testing

  • Storage optimization:

    • Buffer optimization through thermal shift assays

    • Additive screening for stability enhancement

    • Lyophilization protocols for long-term storage

    • Aliquoting to avoid freeze-thaw cycles

These strategies ensure high-quality protein preparations essential for reliable functional and structural studies of B. cereus proteins.

How should researchers interpret contradictory data on B. cereus protein function?

When facing contradictory data on B. cereus protein function, researchers should systematically:

  • Evaluate strain differences:

    • Confirm precise strain identification using MLST

    • Compare virulence gene profiles across experimental strains

    • Consider the 192 different sequence types identified in B. cereus populations

  • Assess experimental conditions:

    • Cell types and their activation states

    • Protein purification methods and potential contaminants

    • Concentration ranges and physiological relevance

    • Temporal factors in experimental design

  • Consider multi-component interactions:

    • For toxins like HBL, ensure all components are present in proper ratios

    • Verify the sequential assembly process is maintained

    • Account for potential co-factors required for activity

  • Examine methodology differences:

    • Direct vs. indirect measurement approaches

    • Sensitivity and specificity of detection methods

    • Potential artifacts from experimental manipulations

  • Resolve contradictions through:

    • Side-by-side comparative studies with standardized protocols

    • Genetic approaches using isogenic mutants (e.g., ΔHbl B. cereus)

    • Complementation studies to restore function

    • Antibody neutralization experiments

  • Incorporate systems biology perspectives:

    • Consider redundancy in biological systems

    • Examine network effects rather than isolated pathways

    • Use multi-omics approaches to capture system-wide effects

The identification of HBL as the inflammasome-activating factor exemplifies this approach, as researchers systematically ruled out NHE and Cytotoxin K despite their similar properties .

What are the critical controls for experiments studying B. cereus protein interactions with host cells?

Rigorous experimental design for B. cereus protein-host cell interaction studies requires comprehensive controls:

  • Bacterial strain controls:

    • Wild-type parental strain

    • Isogenic toxin-deficient mutants (e.g., ΔHbl B. cereus)

    • Complemented mutant strains to restore function

    • Heat-killed bacteria to distinguish active mechanisms from passive recognition

  • Toxin preparation controls:

    • Individual toxin components tested separately

    • Complete toxin complexes in proper ratios

    • Heat-inactivated toxin preparations

    • Antibody-neutralized toxin preparations

    • Contaminant controls (endotoxin-free preparations)

  • Host cell controls:

    • Wild-type cells (e.g., WT BMDMs)

    • Genetically deficient cells (e.g., Nlrp3-/- BMDMs)

    • Pharmacologically inhibited cells (e.g., MCC950-treated cells)

    • Multiple cell types to confirm tissue-specific effects

  • Pathway validation controls:

    • Positive controls for specific pathways (e.g., LPS+ATP for NLRP3 activation)

    • Pathway-independent readouts (e.g., TNF production, IκB and ERK phosphorylation)

    • Dose-response relationships

    • Time-course experiments

  • Technical controls:

    • Vehicle-only treatments

    • Isotype control antibodies

    • Multiple methodological approaches for critical findings

    • Replication across independent experiments

These controls help distinguish specific effects from technical artifacts and establish causality in complex biological systems.

How can structural biology approaches enhance understanding of B. cereus protein functions?

Structural biology provides critical insights into B. cereus protein functions through multiple approaches:

  • X-ray crystallography applications:

    • Determination of high-resolution protein structures

    • Identification of functional domains and active sites

    • Visualization of protein-protein interaction interfaces

    • Co-crystallization with ligands or inhibitors

    • Rational design of inhibitors based on structural features

  • Cryo-electron microscopy advantages:

    • Visualization of large protein complexes (like assembled toxins)

    • Structural determination without crystallization

    • Capturing different conformational states

    • Visualization of membrane insertion for toxins like HBL

    • Structure determination in near-native conditions

  • NMR spectroscopy contributions:

    • Analysis of protein dynamics and conformational changes

    • Characterization of intrinsically disordered regions

    • Direct observation of protein-ligand interactions

    • Study of weak transient interactions

    • Investigation of solution behavior

  • Integrative structural biology:

    • Combining multiple techniques for comprehensive understanding

    • Small-angle X-ray scattering (SAXS) for solution structure

    • Hydrogen-deuterium exchange mass spectrometry for dynamics

    • Computational modeling and simulation to bridge experimental gaps

  • Structure-function correlations:

    • Site-directed mutagenesis based on structural information

    • Functional assays to validate structural hypotheses

    • Engineering of protein variants with altered properties

    • Understanding evolutionary relationships through structural comparison

For multi-component toxins like HBL, structural studies could reveal how the three components (B, L1, L2) assemble sequentially to form functional pores, providing insights that could lead to novel therapeutic approaches .

What bioinformatic approaches are most valuable for predicting B. cereus protein functions?

Bioinformatic analyses provide powerful tools for predicting B. cereus protein functions:

  • Sequence-based predictions:

    • Homology detection using BLAST, HHpred, and HMMER

    • Domain architecture analysis using InterPro and Pfam

    • Motif identification using MEME and PROSITE

    • Signal peptide prediction using SignalP

    • Transmembrane region prediction using TMHMM

  • Structural predictions:

    • Secondary structure prediction (PSIPRED, JPred)

    • Tertiary structure prediction (AlphaFold2, RoseTTAFold)

    • Protein-protein docking (HADDOCK, ClusPro)

    • Molecular dynamics simulations for functional movements

    • Binding site prediction (CASTp, FTMap)

  • Comparative genomics approaches:

    • Phylogenetic profiling to identify functionally related proteins

    • Gene neighborhood analysis for functional associations

    • Analysis of the 192 different sequence types identified in B. cereus

    • Pan-genome analysis to distinguish core vs. accessory genes

    • Detection of horizontal gene transfer events

  • Systems biology integration:

    • Protein-protein interaction network analysis

    • Pathway enrichment analysis

    • Gene co-expression patterns

    • Integration of multi-omics data

    • Text mining of scientific literature

  • Specialized predictions for toxins:

    • Toxic peptide prediction tools

    • Pore-forming motif identification

    • Host-interaction domain prediction

    • Immunogenicity prediction

    • Comparison with known toxin databases

For novel proteins like UPF0145 protein BCE_5284, these approaches can provide initial functional hypotheses to guide experimental design and prioritize research directions.

What are the most effective experimental designs for studying B. cereus protein regulation?

Investigating B. cereus protein regulation requires sophisticated experimental designs:

  • Transcriptional regulation studies:

    • Promoter mapping through 5' RACE

    • Reporter gene assays (luciferase, GFP) with promoter constructs

    • Chromatin immunoprecipitation (ChIP) to identify binding regulators

    • DNA footprinting to precisely locate binding sites

    • Transcription factor identification using pulldown assays

  • Post-transcriptional regulation analysis:

    • RNA stability assessments

    • Translation efficiency studies

    • Identification of small RNAs affecting expression

    • RNA immunoprecipitation to detect RNA-protein interactions

    • Analysis of 5' and 3' UTR regulatory elements

  • Environmental regulation experiments:

    • Controlled exposure to environmental stimuli:

      • Nutrient availability

      • Temperature shifts

      • pH changes

      • Oxygen tension

      • Host factors

    • Real-time PCR for quantitative expression analysis

    • Proteomics to measure resulting protein levels

    • Parallel assessment of multiple virulence factors

  • Genetic approaches:

    • Construction of regulator knockout strains

    • Complementation studies to verify phenotypes

    • Overexpression systems to assess dose effects

    • CRISPR interference for targeted repression

    • Site-directed mutagenesis of regulatory sites

  • Systems-level approaches:

    • RNA-Seq for transcriptome-wide regulation

    • Proteomics for post-translational modification detection

    • Chromatin accessibility mapping

    • Metabolomics to link metabolic state to regulation

    • Network analysis to identify key regulatory nodes

These approaches can reveal how B. cereus regulates toxin expression in response to environmental cues, potentially identifying intervention points for novel therapeutic strategies.

What are the emerging research directions for B. cereus proteins?

Research on B. cereus proteins is evolving toward several promising directions:

  • Host-pathogen interaction characterization: Deeper investigation of how B. cereus toxins like HBL engage with host cellular machinery, particularly the inflammasome pathways that bridge innate immunity and inflammatory disease .

  • Structure-based therapeutic development: Utilizing structural insights into toxin assembly and function to design targeted inhibitors that could neutralize virulence without selecting for antimicrobial resistance .

  • Systems biology integration: Moving beyond single-protein studies to understand how B. cereus toxins and proteins function within comprehensive virulence networks and how these networks respond to environmental signals .

  • Host-directed therapeutic strategies: Building on the finding that NLRP3 inflammasome inhibition with MCC950 prevents B. cereus-induced lethality, suggesting that modulating host response rather than targeting the bacteria directly may be more effective .

  • Personalized infection management: Leveraging the significant genetic diversity observed in B. cereus (192 different sequence types) to develop tailored diagnostic and treatment approaches based on strain-specific virulence profiles .

  • One Health approaches: Integrating human medicine, veterinary science, and environmental health to address B. cereus as a foodborne pathogen that traverses these domains, particularly given its high prevalence (35%) in ready-to-eat foods .

These emerging directions highlight the shift from descriptive to mechanistic understanding and from pathogen-centered to holistic approaches in B. cereus research.

How should researchers integrate B. cereus protein studies into broader microbial pathogenesis frameworks?

Effective integration of B. cereus protein studies into broader pathogenesis frameworks requires:

  • Comparative virulence analysis: Systematically comparing B. cereus virulence mechanisms with related pathogens like B. anthracis and B. thuringiensis to identify conserved and unique pathogenicity strategies.

  • Cross-disciplinary methodology application: Adapting advanced techniques from other fields, such as structural biology, systems biology, and immunology, to illuminate B. cereus pathogenesis.

  • Synthetic biology approaches: Using engineered bacterial systems to reconstitute and study complex virulence mechanisms, such as the sequential assembly of the HBL toxin .

  • Ecological context consideration: Examining how B. cereus proteins function not only in infection but also in environmental persistence, particularly in food matrices where 35% of ready-to-eat samples tested positive .

  • Translational research pathways: Establishing clear connections between basic protein characterization and clinical applications, especially for antimicrobial resistance where nearly 100% of isolates show resistance to penicillin and ampicillin .

  • Multiple model systems utilization: Employing diverse experimental models from cell culture to animal studies to capture the full complexity of protein function in different contexts.

  • Collaborative research frameworks: Forming interdisciplinary teams that combine expertise in protein biochemistry, immunology, microbiology, and clinical research to address the multifaceted nature of B. cereus pathogenesis.

This integrated approach will accelerate discovery and provide more comprehensive understanding of how B. cereus proteins contribute to both pathogenesis and normal bacterial physiology.

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.