KEGG: bca:BCE_4383
The BCE_4383 protein is a probable rRNA maturation factor that plays a critical role in ribosomal RNA processing and maturation in Bacillus cereus. As a member of the Bacillus cereus group, this organism contains various virulence factors and regulatory systems that may interact with or influence the function of BCE_4383 . The protein likely contributes to the organism's ability to adapt to various environmental conditions, similar to other regulatory proteins in the B. cereus group. The function of this protein must be understood within the context of B. cereus' lifecycle, which includes both vegetative growth and sporulation phases .
For optimal expression of recombinant BCE_4383 in E. coli systems, researchers should consider the following protocol:
Vector selection: pET expression systems with T7 promoters typically yield high expression levels for Bacillus proteins.
E. coli strain: BL21(DE3) or Rosetta strains are recommended for enhanced expression of proteins with rare codons.
Induction parameters:
Temperature: 18-25°C for expression after induction (lower temperatures reduce inclusion body formation)
IPTG concentration: 0.1-0.5 mM
Duration: 16-18 hours for optimal protein folding
Based on protocols established for similar Bacillus proteins, transformation can follow standard methods similar to those used for B. cereus plasmid transformation, where approximately 1 μg of plasmid DNA is mixed with competent cells and subjected to electroporation (0.6 kV, 500 Ω, and 25 μF) . After expression, purification typically employs affinity chromatography followed by size exclusion methods to ensure protein integrity.
CRISPR/Cas9 genome editing has proven highly effective for genetic manipulation in the Bacillus cereus group and can be applied to study BCE_4383 function through the following approach:
Design of sgRNA: Target sequences specific to BCE_4383 with minimal off-target effects, optimally 20 nucleotides in length with an adjacent PAM sequence (NGG).
Vector construction: Develop an all-in-one CRISPR-Cas9 plasmid containing:
Cas9 gene under mannose-inducible promoter
sgRNA expression cassette
Homologous arms (500-1000 bp each) flanking the target site for gene deletion or mutation
Transformation protocol:
Induction and selection:
Verification: Confirm successful editing by PCR amplification and sequencing of the target region.
This method has demonstrated high efficiency in generating precise mutations in B. cereus genes, with success rates comparable to those achieved in B. anthracis genome modifications .
When designing experiments to assess BCE_4383 interactions with ribosomes, researchers should implement the following comprehensive approach:
Experimental design principles:
In vitro binding assays:
Ribosome isolation: Purify intact ribosomes or ribosomal subunits from B. cereus using differential centrifugation
Protein labeling: Employ fluorescent tags or radioactive isotopes that minimally interfere with protein function
Binding conditions: Test multiple buffer compositions to identify optimal interaction conditions
Detection methods: Employ filter binding assays, surface plasmon resonance, or microscale thermophoresis for quantitative measurement
Structural analysis:
Cryo-EM studies of BCE_4383-ribosome complexes at various assembly stages
Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces
Crosslinking followed by mass spectrometry to identify specific contact residues
Functional assays:
In vitro transcription-translation systems to assess the impact on protein synthesis
Ribosome assembly assays to determine the stage at which BCE_4383 acts
Competitive binding with known rRNA maturation factors to establish hierarchy
Data analysis strategy:
Determine binding kinetics parameters (Kd, kon, koff)
Apply statistical methods appropriate for multiple variable analysis
Use computational modeling to integrate experimental findings
Mutations in BCE_4383 may significantly impact B. cereus virulence through alterations in ribosome biogenesis and protein synthesis efficiency. A systematic approach to investigate this relationship would include:
Creation of mutant strains:
Generate point mutations or domain deletions using CRISPR/Cas9 genome editing
Develop complementation strains expressing wild-type BCE_4383 to verify phenotype rescue
Create reporter strains with fluorescently tagged virulence factors to monitor expression
Virulence factor expression analysis:
Infection models:
Cellular models: Assess cytotoxicity against human epithelial cells and macrophages
Animal models: Evaluate bacterial load, dissemination, and host survival in appropriate models
Tissue-specific studies: Examine effects on pulmonary, ocular, or CNS infections based on B. cereus' known pathogenic profile
Comparative analysis with clinical isolates:
Sequence BCE_4383 in clinical isolates from various infection types
Correlate sequence variations with virulence phenotypes and clinical outcomes
Develop a predictive model of mutation impact on pathogenicity
B. cereus is known to cause various systemic and local infections, including fulminant bacteremia, CNS involvement, endophthalmitis, pneumonia, and cutaneous infections . Understanding how BCE_4383 mutations affect these pathogenic capabilities would provide valuable insights into B. cereus virulence mechanisms and potential therapeutic targets.
The probable rRNA maturation factor BCE_4383 likely plays a crucial role in B. cereus adaptation to environmental stresses through modulation of ribosome assembly and function. To investigate this relationship:
Stress exposure studies:
Subject wild-type and BCE_4383 mutant strains to various stresses:
Heat shock (42-45°C)
Oxidative stress (H₂O₂, paraquat)
Antibiotic exposure (particularly those targeting translation)
Nutrient limitation
pH extremes
Quantitative assessment methods:
Growth kinetics analysis under various stress conditions
Measurement of survival rates following acute stress exposure
Determination of minimum inhibitory concentrations for various antibiotics
Assessment of spore formation efficiency and germination rates
Molecular response characterization:
Ribosome profiling to assess translation dynamics under stress
Quantification of rRNA processing intermediates
ChIP-seq analysis to identify stress-induced changes in BCE_4383 binding patterns
Protein-protein interaction studies to identify stress-specific binding partners
Transcriptional regulation:
Analyze BCE_4383 expression under different stress conditions
Identify transcription factors regulating BCE_4383 expression
Examine cross-talk with known stress response regulators
This approach would elucidate how BCE_4383 contributes to the remarkable adaptability of B. cereus to diverse environmental conditions and potentially explain its ubiquitous distribution in soil, food, and marine environments .
The relationship between BCE_4383 activity and sporulation in B. cereus represents a critical aspect of this organism's lifecycle. To investigate this connection:
Temporal expression analysis:
Monitor BCE_4383 expression throughout the sporulation process using:
Transcriptomics (RNA-seq or qRT-PCR)
Western blotting with specific antibodies
Fluorescent protein fusions to track localization
Genetic manipulation approaches:
Create conditional BCE_4383 mutants using inducible promoters
Analyze sporulation efficiency in BCE_4383 deletion/depletion strains
Assess the impact of BCE_4383 overexpression on sporulation timing and efficiency
Microscopy-based assessment:
Employ phase-contrast microscopy to monitor morphological changes
Use fluorescence microscopy with membrane and DNA stains to track sporulation stages
Implement time-lapse microscopy to capture the entire sporulation process
Biochemical characterization:
Analyze rRNA processing patterns during sporulation in wild-type vs. mutant strains
Assess ribosome profiles during transition from vegetative growth to sporulation
Identify sporulation-specific interaction partners of BCE_4383
Comparative analysis with other Bacillus species:
Compare BCE_4383 function in B. cereus with homologs in B. anthracis and B. thuringiensis
Correlate differences in sporulation properties with BCE_4383 sequence variations
This investigation would provide insights into how BCE_4383 contributes to the sporulation process that is central to B. cereus persistence in diverse environments and its ability to cause food poisoning and other infections .
Researchers frequently encounter several challenges when purifying recombinant BCE_4383. Here are systematic approaches to address these issues:
Poor solubility and inclusion body formation:
Solution: Optimize expression conditions by reducing temperature (16-20°C), lowering inducer concentration, and using slower induction
Alternative: Express as a fusion protein with solubility-enhancing tags (MBP, SUMO, or TrxA)
Recovery method: If inclusion bodies persist, develop a refolding protocol using step-wise dialysis with decreasing concentrations of chaotropic agents
Proteolytic degradation:
Solution: Add protease inhibitor cocktails during all purification steps
Alternative: Co-express with chaperones to enhance proper folding
Optimization: Identify and mutate protease-sensitive sites without affecting protein function
Co-purification of nucleic acids:
Solution: Increase salt concentration (up to 1M NaCl) in lysis and wash buffers
Treatment: Add nucleases (DNase I, RNase A) during initial purification steps
Validation: Monitor A260/A280 ratio to ensure protein purity
Loss of activity during purification:
Stabilization: Include specific cofactors or metal ions required for proper folding
Buffer optimization: Test various buffer compositions (HEPES, Tris, phosphate) and pH values
Storage: Determine optimal storage conditions (glycerol percentage, temperature, additives)
Purification troubleshooting table:
| Issue | Possible Cause | Solution | Validation Method |
|---|---|---|---|
| Low yield | Poor expression | Optimize codon usage, change expression strain | SDS-PAGE analysis |
| Multiple bands | Degradation | Add protease inhibitors, reduce purification time | Western blot |
| Inactive protein | Improper folding | Include chaperones, optimize refolding | Activity assay |
| Aggregation | Hydrophobic interactions | Add mild detergents, increase salt | Dynamic light scattering |
| Contaminants | Non-specific binding | Increase imidazole in wash buffers | Mass spectrometry |
These methodological approaches are based on established protocols for purifying recombinant proteins from Bacillus species and can be adapted specifically for BCE_4383 based on its unique characteristics.
Distinguishing between direct effects of BCE_4383 mutation and polar effects on adjacent genes requires a systematic experimental approach:
Complementation strategies:
In trans complementation: Introduce wild-type BCE_4383 on a plasmid under native or inducible promoter
Chromosomal complementation: Reintroduce BCE_4383 at a neutral site in the chromosome
Dose-dependent complementation: Use various promoter strengths to assess the relationship between BCE_4383 expression levels and phenotype restoration
Targeted mutagenesis approaches:
Silent mutations: Introduce synonymous mutations that maintain protein sequence while altering nucleotide sequence
Domain-specific mutations: Target specific functional domains rather than creating complete gene deletions
CRISPR interference (CRISPRi): Use catalytically inactive Cas9 for targeted gene repression without genomic alterations
Transcriptional analysis:
Strand-specific RNA-seq: Assess impact on adjacent gene expression
qRT-PCR validation: Quantify expression changes in genes upstream and downstream
Promoter fusion studies: Create transcriptional fusions to report on promoter activity
Operon structure analysis:
5' RACE: Define transcriptional start sites of adjacent genes
RT-PCR spanning gene junctions: Determine if BCE_4383 is part of an operon
Northern blot analysis: Identify transcript sizes to determine co-transcription
The CRISPR/Cas9 system described for B. cereus can be particularly useful here, as it allows for precise genome editing with minimal disruption to surrounding sequences . By designing homologous arms that precisely delete only the target gene while maintaining the integrity of adjacent regulatory sequences, researchers can minimize polar effects and create clean deletions for phenotypic analysis.
Crystallizing BCE_4383 for structural studies presents several challenges that can be addressed through the following systematic approaches:
Protein sample optimization:
Construct design: Create multiple constructs with different N- and C-terminal boundaries to remove flexible regions
Surface engineering: Introduce surface entropy reduction mutations (replacing clusters of high-entropy residues like Lys, Glu, and Gln with alanines)
Fusion partners: Utilize crystallization chaperones such as T4 lysozyme, MBP, or BRIL to provide crystal contacts
Crystallization condition screening:
Initial screening: Employ sparse matrix screens covering diverse precipitants, buffers, and additives
Grid screening: Optimize promising conditions by systematically varying pH, precipitant concentration, and temperature
Microseeding: Introduce crystal seeds into fresh drops to promote nucleation and growth
Alternative approaches: Consider counter-diffusion, batch, or dialysis methods if vapor diffusion is unsuccessful
Complex formation strategies:
Ligand co-crystallization: Include substrate analogs, inhibitors, or binding partners
Stabilizing agents: Add small molecules that enhance conformational stability
Metal ions: Screen various metal ions that might be critical for structural integrity
Alternative structural approaches:
Cryo-electron microscopy: For BCE_4383-ribosome complexes or if crystal formation remains challenging
NMR spectroscopy: For structural analysis of isolated domains
Small-angle X-ray scattering (SAXS): For low-resolution envelope determination and conformational dynamics
Crystallization optimization table:
| Parameter | Variation Range | Optimization Strategy |
|---|---|---|
| Protein concentration | 2-20 mg/ml | Systematic screening with 2 mg/ml increments |
| Temperature | 4°C, 16°C, 20°C | Parallel trials at different temperatures |
| Precipitant concentration | 50-150% of initial hit | Fine gradient around promising conditions |
| Additive screening | Detergents, alcohols, polyamines | Supplement base conditions with commercial additive screens |
| Drop ratio (protein:reservoir) | 1:1, 1:2, 2:1 | Vary drop composition to alter equilibration rate |
These approaches have proven successful for crystallizing challenging bacterial proteins, including those involved in RNA processing and maturation.
High-throughput screening (HTS) approaches for identifying BCE_4383 inhibitors as potential antimicrobial agents could follow this comprehensive workflow:
Assay development and validation:
Primary assay: Develop a fluorescence-based or bioluminescent assay measuring BCE_4383 activity on model RNA substrates
Counter-screening: Establish assays to eliminate compounds that non-specifically interfere with RNA or general RNA-binding proteins
Validation criteria: Z' factor > 0.5, signal-to-background ratio > 10, coefficient of variation < 10%
Compound library selection:
Diverse chemical libraries: Begin with 50,000-100,000 compounds covering broad chemical space
Focused libraries: Include known RNA-binding compounds, nucleoside analogs, and antimicrobials
Natural product extracts: Screen fractionated extracts from microorganisms and plants
Screening cascade:
Primary screen: Test compounds at a single concentration (10-20 μM)
Dose-response confirmation: Generate full dose-response curves for hits (IC₅₀ determination)
Secondary assays: Validate hits in orthogonal biochemical assays
Cellular validation: Assess antimicrobial activity against B. cereus and related pathogens
Hit-to-lead optimization:
Structure-activity relationship studies: Synthesize analogs to improve potency and selectivity
Mode of action studies: Determine binding site through mutagenesis and structural studies
ADME-Tox assessment: Evaluate drug-like properties and potential toxicity
In vivo efficacy: Test lead compounds in relevant infection models
Key success criteria for inhibitor development:
| Parameter | Threshold for Progression | Method of Determination |
|---|---|---|
| BCE_4383 inhibition | IC₅₀ < 1 μM | Enzymatic assay |
| Antimicrobial activity | MIC < 8 μg/ml against B. cereus | Broth microdilution |
| Selectivity | >50-fold vs. human homologs | Comparative biochemical assays |
| Cytotoxicity | CC₅₀ > 50 μM in mammalian cells | MTT or similar viability assay |
| Solubility | >100 μM in aqueous solution | Kinetic solubility assay |
Given B. cereus' role as a significant human pathogen causing food poisoning, systemic infections, and even anthrax-like progressive pneumonia , developing specific inhibitors targeting BCE_4383 could provide valuable new therapeutic approaches, especially for infections resistant to conventional antibiotics.
Comparative genomics approaches can provide significant insights into the evolution of BCE_4383 across the Bacillus cereus group through the following multifaceted analysis:
Phylogenetic analysis:
Sequence alignment: Construct multiple sequence alignments of BCE_4383 homologs from all sequenced B. cereus group species
Tree construction: Generate phylogenetic trees using maximum likelihood and Bayesian methods
Evolutionary rate analysis: Calculate dN/dS ratios to identify sites under positive or purifying selection
Structural conservation assessment:
Domain architecture: Identify conserved and variable domains across species
Functional site prediction: Map conserved residues onto structural models
Comparative structural modeling: Generate models for BCE_4383 variants across species
Genomic context analysis:
Synteny mapping: Examine conservation of gene neighborhoods around BCE_4383
Operon structure comparison: Determine if operon organization varies between species
Mobile genetic element association: Identify any association with transposons, phages, or other mobile elements
Functional divergence investigation:
Expression pattern comparison: Analyze transcriptomic data across species under various conditions
Regulatory element identification: Compare promoter regions and transcription factor binding sites
Post-translational modification prediction: Identify potential species-specific modifications
Ecological and pathogenic correlation:
Niche adaptation: Correlate BCE_4383 sequence variations with ecological niches
Virulence association: Examine relationships between BCE_4383 variants and pathogenic potential
Host range determination: Assess if BCE_4383 variations correlate with host specificity
This comparative approach would be particularly informative given the close genetic relationship but distinct ecological and pathogenic characteristics of the B. cereus group members, which includes the highly pathogenic B. anthracis alongside less virulent strains . Understanding the evolution of BCE_4383 could provide insights into how rRNA processing machinery has adapted across this diverse bacterial group and potentially reveal novel targets for species-specific interventions.
Effective collaboration on BCE_4383 studies across specialties and institutions requires a structured approach:
Interdisciplinary research framework:
Core expertise integration: Combine molecular biologists, structural biologists, microbiologists, and bioinformaticians
Technology platform sharing: Establish protocols for sharing specialized equipment and methodologies
Complementary skill mapping: Identify unique capabilities at each institution to maximize resource utilization
Standardized protocols and data sharing:
Method harmonization: Develop and validate consistent experimental protocols across sites
Data repository: Establish a centralized database for raw data, results, and analyses
Material exchange: Create a standardized system for sharing strains, plasmids, and reagents
Communication infrastructure:
Regular virtual meetings: Schedule recurring video conferences with defined agendas
Progress tracking system: Implement project management software to monitor milestones
Real-time collaboration tools: Utilize electronic lab notebooks and collaborative document platforms
Integrated experimental design:
Design of experiments approach: Apply factorial experimental design to systematically address complex questions
Sequential validation: Design studies where results from one institution inform experiments at another
Complementary methodologies: Apply different techniques to answer the same research question
Collaborative publication strategy:
Authorship guidelines: Establish clear criteria for authorship and author order
Manuscript development workflow: Create a structured process for manuscript preparation with defined responsibilities
Preprint sharing: Utilize preprint servers to gather community feedback before formal submission
This collaborative framework would be particularly valuable for BCE_4383 research given the multifaceted nature of rRNA maturation factors and the diverse techniques required to fully characterize their function, from genomic manipulation using CRISPR/Cas9 systems to structural and biochemical analyses.