Recombinant Bacillus subtilis uncharacterized membrane protein ylmG (ylmG) is a partial recombinant protein derived from the ylmG gene (UniProt: O31729) of Bacillus subtilis (strain 168). While its precise biological function remains uncharacterized, it is classified as a membrane-associated protein based on sequence predictions . The recombinant form is commercially available for research purposes, produced in yeast systems, and purified to >85% purity via SDS-PAGE .
Partial Sequence: The recombinant product includes a truncated version of the native protein. Full-length ylmG is predicted to contain transmembrane domains, though specific topology remains unconfirmed .
Homology: Limited functional homologs identified in public databases, emphasizing its classification as an "uncharacterized" protein .
Recombinant ylmG is synthesized in yeast via heterologous expression systems. Post-production steps include:
Purification: Immobilized metal affinity chromatography (IMAC) for His-tagged variants .
Quality Control: SDS-PAGE validation to ensure >85% purity .
Reconstitution: Recommended in deionized sterile water with 50% glycerol for long-term stability .
While ylmG’s role is unknown, its classification as a membrane protein positions it within broader studies of B. subtilis membrane protein biogenesis. Key insights from related systems include:
YidC/Oxa1/Alb3 Family: B. subtilis employs YidC1 (spoIIIJ) and YidC2 (yqjG) for membrane protein insertion. MifM, a regulatory nascent chain, senses YidC1 activity and induces YidC2 expression when insertion capacity is limited .
Dynamic Localization: Membrane proteins in B. subtilis exhibit non-random distribution, with domains forming discrete clusters (e.g., ATP synthase, succinate dehydrogenase) .
Host Advantages: B. subtilis is a GRAS-certified organism with robust secretion systems (Sec and Tat pathways), enabling high-yield recombinant protein production .
Challenges: Protein misfolding and proteolytic degradation remain bottlenecks, necessitating optimized signal peptides and chaperone systems .
Functional Role: No studies directly link ylmG to known pathways (e.g., stress response, nutrient uptake).
Structural Insights: Crystallization or cryo-EM data are absent, limiting mechanistic understanding.
Interaction Partners: Potential interactions with YidC, MifM, or other membrane proteins remain unexplored .
KEGG: bsu:BSU15400
STRING: 224308.Bsubs1_010100008516
The ylmG protein is an uncharacterized membrane protein in Bacillus subtilis. While its precise function remains to be fully elucidated, it belongs to the category of bacterial membrane proteins that potentially play roles in cellular processes including membrane organization, transport, signaling, and cellular division. The significance of studying ylmG extends beyond understanding its specific function to developing broader insights into bacterial membrane protein biology. As an uncharacterized protein, ylmG research contributes to filling knowledge gaps in bacterial proteomes and potentially uncovering novel cellular mechanisms. The protein's membrane localization makes it particularly valuable for studying membrane protein biogenesis, topology, and function in Gram-positive bacteria.
For studying recombinant ylmG in B. subtilis, several expression systems can be employed based on research objectives. B. subtilis offers significant advantages as both a host and source organism due to its GRAS (Generally Recognized As Safe) status and remarkable innate ability to absorb and incorporate exogenous DNA into its genome . For membrane proteins like ylmG, expression systems using inducible promoters such as Pspac (IPTG-inducible) or PxylA (xylose-inducible) are commonly recommended.
Expression System | Inducer | Advantages | Considerations for ylmG |
---|---|---|---|
Pspac system | IPTG | Tight regulation, dose-dependent expression | May require optimization for membrane protein expression |
PxylA system | Xylose | Lower basal expression | Good for potentially toxic membrane proteins |
Phyper-spank | IPTG | High expression levels | May lead to inclusion bodies for membrane proteins |
Self-inducible systems | Auto-induction | Simplified cultivation | May affect membrane integrity |
When selecting an expression system, it's crucial to consider that the genetic engineering strategy should accommodate the hydrophobic nature of membrane proteins like ylmG, potentially incorporating signal peptides for proper membrane integration .
Studying uncharacterized membrane proteins like ylmG presents several key methodological challenges:
Protein expression and solubilization: Membrane proteins often express at low levels and can be difficult to extract from membranes while maintaining native conformation. This requires careful optimization of expression conditions and solubilization methods.
Protein purification: The hydrophobic nature of membrane proteins makes them challenging to purify without aggregation, necessitating specialized detergent screening and purification protocols.
Structural characterization: Obtaining high-resolution structural data requires specialized techniques adapted for membrane proteins, which are inherently more challenging than soluble proteins.
Functional assignment: Without known homologs or established functional assays, determining the function of ylmG requires multiple complementary approaches.
Localization confirmation: Verifying proper membrane integration and determining topology is essential for functional studies but requires specialized techniques.
These challenges are typically addressed through a combination of genetic fusion approaches (such as GFP tagging), optimized detergent screening, and employing multiple complementary characterization techniques. The technological arsenal available for B. subtilis expression platforms continues to improve, allowing for more efficient production of membrane proteins of biotechnological importance .
Determining membrane topology of ylmG requires a methodical experimental design approach integrating computational and experimental methods:
Computational prediction: Begin with in silico analysis using tools like TMHMM, Phobius, or TOPCONS to predict transmembrane domains and orientation. This provides initial hypotheses about membrane-spanning regions and their orientation.
Fusion protein approach: Create systematic fusions with reporter proteins such as:
PhoA (alkaline phosphatase): Active only in periplasm/extracellular space
GFP: Fluorescent only when properly folded in cytoplasm
LacZ (β-galactosidase): Active only in cytoplasm
Cysteine accessibility method: Introduce cysteine residues at predicted loops, then test accessibility with membrane-permeable and impermeable thiol-reactive reagents to determine which regions are exposed to each side of the membrane.
Protease protection assays: Expose membrane preparations to proteases with/without membrane disruption to identify protected domains, providing information about which regions are accessible.
Data integration is critical - concordance between multiple methods provides stronger evidence for proposed topology models. For ylmG specifically, using insights from experimental design for big data analysis can help optimize the number of positions tested while maximizing information gain .
When investigating interaction partners of an uncharacterized membrane protein like ylmG, robust controls are essential for reliable results:
Positive controls:
Known membrane protein interaction pairs in B. subtilis
Artificially constructed interacting membrane proteins
If available, homologous protein interactions from related species
Negative controls:
Empty vector constructs
Non-interacting membrane proteins
Scrambled/mutated ylmG sequences that should disrupt interactions
Methodology controls:
Input protein quantification
Reverse pull-down experiments (bait-prey reversal)
Competition assays with unlabeled proteins
Detergent controls to rule out non-specific hydrophobic interactions
For biological validation, complementary methods should be employed following principles of optimal experimental design :
Co-immunoprecipitation or pull-down assays
Bacterial two-hybrid assays adapted for membrane proteins
FRET or BiFC for in vivo validation
Cross-linking followed by mass spectrometry
Data should be presented as normalized interaction strength relative to controls, with statistical analysis of replicate experiments. This multi-modal approach provides higher confidence in true interaction partners versus false positives.
Optimizing recombinant ylmG expression for structural studies requires systematic optimization across multiple parameters:
Expression construct design:
Expression conditions optimization:
Host strain selection:
Test protease-deficient strains
Consider strains with altered membrane composition
Evaluate strains overexpressing chaperones
Scale-up strategy:
Bioreactor cultivation with controlled dissolved oxygen
Fed-batch approaches to maximize biomass
Induction protocols optimized for membrane protein expression
Optimization matrix for ylmG expression:
Parameter | Variables to test | Expected impact | Measurement metric |
---|---|---|---|
Temperature | 16°C, 25°C, 30°C | Lower temp may improve folding | Western blot, membrane fraction yield |
Induction time | Early log, mid-log, late log | Phase-dependent expression | Protein yield per cell mass |
Inducer concentration | 0.1-1.0 mM IPTG or 0.1-2% xylose | Optimal induction level | Total protein yield, soluble fraction yield |
Media supplements | Glycerol, betaine, sucrose | Osmolyte-assisted folding | Functional protein yield |
This systematic optimization approach applies principles from experimental design theory to efficiently identify optimal conditions through strategic sampling of the parameter space .
Confirming subcellular localization of ylmG requires multiple complementary approaches:
Fluorescence microscopy techniques:
C-terminal or internal GFP fusions (confirming function is maintained)
Immunofluorescence with antibodies against ylmG or epitope tags
Super-resolution techniques (STED, PALM, STORM) for precise localization
Co-localization studies with known membrane compartment markers
Biochemical fractionation:
Differential centrifugation to separate cellular compartments
Density gradient fractionation for membrane separation
Western blotting of fractions with compartment-specific controls
Detection using optimized antibodies or epitope tags
Protease accessibility assays:
Selective permeabilization of cellular compartments
Proteinase K treatment of intact cells vs. spheroplasts
Analysis of protected fragments
Electron microscopy approaches:
Immunogold labeling with ylmG-specific antibodies
Cryo-electron microscopy of membrane preparations
For ylmG, a comprehensive analysis should include quantification of co-localization coefficients with known membrane markers and statistical analysis of spatial distribution patterns across growth phases. Results can be compared with localization patterns of established membrane proteins in B. subtilis to provide context for interpretation.
For identifying post-translational modifications (PTMs) of ylmG, several mass spectrometry approaches are recommended:
Sample preparation strategies:
Enrichment of modified peptides (IMAC for phosphorylation, lectin affinity for glycosylation)
Multiple protease digestions to optimize sequence coverage
Specialized extraction protocols for membrane proteins
MS methodologies:
Bottom-up proteomics: Tryptic digestion followed by LC-MS/MS
Top-down proteomics: Analysis of intact protein
Middle-down approach: Limited digestion to generate larger peptide fragments
Fragmentation techniques:
CID (collision-induced dissociation): Standard approach
ETD/ECD (electron transfer/capture dissociation): Better for labile modifications
HCD (higher-energy collisional dissociation): Improved fragment detection
Data analysis workflow:
Database searching with variable modification options
De novo sequencing for unexpected modifications
Site localization scoring algorithms
Manual verification of key spectra
The most common PTMs to investigate for bacterial membrane proteins like ylmG include phosphorylation, methylation, and lipid modifications. For each identified modification, validation through site-directed mutagenesis and functional assays is recommended to establish biological significance.
Resolving contradictory structural predictions for ylmG requires a systematic approach combining computational and experimental validation:
Comparative analysis of prediction methods:
Create a consensus table of predictions from multiple tools
Weight predictions based on algorithm performance for membrane proteins
Identify regions of agreement and disagreement
Experimental validation strategies:
Target contradictory regions for focused experimental validation
Use orthogonal experimental approaches like:
Cysteine scanning mutagenesis
Trp fluorescence quenching
FRET-based distance measurements
Disulfide cross-linking of predicted proximal residues
Homology-based assessment:
Evaluate conservation patterns in homologous proteins
Apply evolutionary coupling analysis to identify co-evolving residues
Use any available structures of distant homologs as templates
Integrative modeling approach:
Combine computational predictions with experimental constraints
Implement Bayesian statistical frameworks to weight conflicting data
Generate ensemble models that capture structural uncertainty
Decision matrix for resolving structural contradictions:
Contradiction type | Primary validation method | Secondary validation | Confidence metric |
---|---|---|---|
TM helix boundaries | Glycosylation mapping | Cys accessibility | Agreement between ≥2 methods |
Cytoplasmic vs. periplasmic loops | PhoA/GFP fusions | Antibody accessibility | Statistical significance of activity ratios |
Secondary structure elements | CD spectroscopy | HDX-MS | Consensus of ≥3 prediction tools |
Tertiary contacts | Cross-linking | Double-mutant cycles | Reproducibility across conditions |
This approach follows principles of optimal experimental design by strategically targeting high-information regions of uncertainty rather than exhaustively testing all possibilities .
Designing a CRISPR-Cas9 approach for studying ylmG requires careful consideration of B. subtilis-specific parameters:
CRISPR system selection:
Implement a codon-optimized Cas9 for B. subtilis
Consider alternative Cas variants (Cas12a/Cpf1) for different PAM requirements
Evaluate catalytically dead Cas9 (dCas9) for CRISPRi gene repression
gRNA design strategy:
Target early in coding sequence for gene disruption
Design multiple gRNAs to minimize off-target effects
For CRISPRi, target near promoter or early in coding sequence
Confirm PAM site availability in ylmG sequence
Delivery and expression:
Integrate Cas9 into chromosome or use plasmid-based expression
Implement inducible promoters for controlled Cas9 expression
Design temperature-sensitive plasmids for transient expression
Editing strategies:
Gene disruption: Introduce frameshift mutations
Precise editing: Provide repair templates with desired modifications
Functional domain analysis: Create truncations or domain deletions
Tagged variants: Introduce epitope tags or fluorescent proteins
Phenotypic analysis:
CRISPR experimental design table for ylmG functional analysis:
Objective | CRISPR approach | Repair template | Validation method | Expected outcome |
---|---|---|---|---|
Complete knockout | gRNA targeting early exon | None (NHEJ repair) | PCR, sequencing, Western blot | Loss of protein, phenotype assessment |
Domain disruption | Multiple gRNAs targeting functional regions | HDR templates with stop codons | Domain-specific antibodies, functional assays | Domain-specific functional insights |
Tagged variant | gRNA near terminus | HDR template with tag sequence | Fluorescence, pull-down assays | Localization and interaction studies |
Conditional knockdown | dCas9 with promoter-targeting gRNA | N/A | RT-qPCR, Western blot | Titratable reduction in expression |
High-throughput approaches for studying condition-dependent ylmG expression or localization include:
Transcriptional analysis:
RNA-Seq under diverse conditions (stress, growth phases, nutrients)
Promoter-reporter fusions in microplate format
Tiling array analysis for transcriptional start site identification
ChIP-Seq to identify regulatory proteins binding near ylmG
Translational and post-translational monitoring:
Ribosome profiling across conditions
MS-based proteomics with SILAC or TMT labeling
Pulse-chase experiments with automated sampling
High-content microscopy of fluorescently tagged ylmG
Phenotypic screening:
Data integration approaches:
Machine learning algorithms to identify condition-dependent patterns
Network analysis to place ylmG in regulatory contexts
Comparative genomics across Bacillus species
Experimental matrix for condition screening:
Environmental factor | Parameter range | Measurement approach | Data analysis method |
---|---|---|---|
Temperature | 15-45°C, 5°C intervals | Fluorescence microscopy, Western blot | Quantitative image analysis, expression normalization |
Osmotic stress | 0-1.5M NaCl | Time-lapse microscopy, fractionation | Localization change kinetics |
Cell wall stress | Various antibiotics | RNA-Seq, proteomics | Differential expression analysis |
Growth phase | Early log to stationary | Ribosome profiling, MS | Time-series clustering |
Nutrient limitation | C, N, P starvation | ChIP-Seq, metabolomics | Regulatory network modeling |
This approach applies principles of experimental design for big data analysis, using systematic sampling of conditions to maximize information gain while minimizing experimental effort .
Integrating computational and experimental approaches for ylmG functional prediction requires a multi-layered strategy:
Computational prediction pipeline:
Sequence-based analysis: PSI-BLAST, HMMer for distant homologs
Structure prediction: AlphaFold2, RoseTTAFold for 3D modeling
Domain and motif identification: InterPro, PFAM, PROSITE
Genomic context: Gene neighborhood conservation, operon analysis
Co-evolution analysis: Direct coupling analysis, mutual information
Targeted experimental validation:
Site-directed mutagenesis of predicted functional residues
Heterologous expression to test functional complementation
Protein-protein interaction studies based on predicted partners
Substrate screening based on structural binding pocket analysis
Iterative refinement:
Update computational models with experimental results
Employ Bayesian approaches to integrate diverse data types
Develop machine learning models trained on validated features
Functional assignment frameworks:
Gene Ontology enrichment of network neighbors
Phylogenetic profiling correlation analysis
Metabolic network gap analysis
Phenotypic clustering of similar mutants
Integrated function prediction workflow:
Stage | Computational methods | Experimental validation | Integration approach |
---|---|---|---|
Initial prediction | Homology modeling, domain analysis | Localization, topology mapping | Structural constraint refinement |
Interaction partners | Docking simulations, co-evolution | Y2H/BACTH, pull-downs, BiFC | Network analysis |
Functional context | Pathway mapping, gene neighborhood | Growth phenotypes, metabolomics | Pathway gap analysis |
Mechanism hypothesis | MD simulations, QM/MM | Site-directed mutagenesis, activity assays | Mechanistic modeling |
This integrated approach applies principles from experimental design theory to efficiently allocate resources between computational and experimental methods, maximizing information gain .
Troubleshooting undetectable ylmG expression requires systematic investigation of multiple potential failure points:
Transcriptional issues:
Verify promoter functionality with a reporter gene
Check for mutations in promoter region by sequencing
Assess transcription by RT-PCR or Northern blot
Consider cryptic regulatory elements affecting expression
Translation problems:
Analyze codon usage optimization for B. subtilis
Check for rare codons that might stall translation
Verify ribosome binding site integrity and spacing
Consider mRNA secondary structure impeding translation initiation
Protein stability issues:
Test for rapid protein degradation using protease inhibitors
Create fusion with stabilizing partners
Evaluate growth temperature effects on stability
Consider toxicity leading to selection against expressing cells
Detection limitations:
Verify antibody specificity and sensitivity
Try alternative tags for detection
Enrich membrane fractions before analysis
Use more sensitive detection methods (e.g., MS instead of Western blot)
B. subtilis has a remarkable ability to absorb and incorporate exogenous DNA, but expression of recombinant membrane proteins presents unique challenges . Several genetic engineering strategies may need to be explored, including different plasmids, promoters, and secretion systems to achieve detectable expression.
Distinguishing true functional localization from non-specific membrane association requires multiple lines of evidence:
Membrane specificity analysis:
Compare localization across different membrane fractions
Use density gradient separation of membrane types
Employ lipid-specific dyes for co-localization studies
Test localization in membrane-composition mutants
Dynamics assessment:
FRAP (Fluorescence Recovery After Photobleaching) to measure mobility
Single-molecule tracking to analyze diffusion coefficients
Inducible expression systems to monitor de novo localization
Functional perturbation:
Site-directed mutagenesis of putative targeting sequences
Domain deletion analysis to identify localization determinants
Heterologous expression in different bacterial species
Competition with overexpressed targeting domains
Co-localization with functional markers:
Dual-color imaging with known membrane domain markers
Quantitative co-localization analysis (Pearson's coefficient, Manders' overlap)
Proximity ligation assays with potential interaction partners
Spatiotemporal correlation with cellular processes
Analytical framework for assessing specific localization:
Test | Non-specific association | Functional localization | Quantitative metric |
---|---|---|---|
Membrane fractionation | Present in all membrane fractions | Enriched in specific fractions | Enrichment factor >3 |
FRAP analysis | Rapid, complete recovery | Slower, potentially incomplete recovery | Recovery half-time, immobile fraction |
Mutation effects | Minimal impact of point mutations | Specific mutations abolish localization | Correlation with functional impact |
Competition assays | Easily displaced by non-specific factors | Only displaced by specific competitors | IC50 values of competitors |
Similar approaches have been used to study functional localization of other B. subtilis proteins involved in processes like DNA damage response .
Resolving conflicting phenotypic data from different ylmG mutant strains requires systematic analysis:
Genetic background verification:
Whole genome sequencing to identify compensatory mutations
Backcross mutants to wild-type to eliminate secondary mutations
Construct clean deletions/mutations in multiple reference strains
Create merodiploid strains to test dominance relationships
Methodological standardization:
Standardize growth conditions, media preparation, and cell handling
Implement blinded analysis of phenotypes to reduce bias
Quantitative rather than qualitative phenotypic measurements
Statistical power analysis to determine appropriate sample sizes
Phenotypic spectrum analysis:
Create allelic series of mutations (null, hypomorph, separation-of-function)
Test phenotypes across varied conditions (temperature, stress, nutrients)
Time-resolved phenotypic analysis throughout growth phases
Single-cell analysis to identify population heterogeneity, similar to approaches used for DNA damage response studies
Data integration approaches:
Principal component analysis of multi-parametric phenotypes
Hierarchical clustering of mutants based on phenotypic profiles
Bayesian networks to identify causal relationships
Meta-analysis methodologies to integrate disparate datasets
Taking inspiration from studies on B. subtilis DNA damage response proteins, measuring cell length distributions across different conditions and treatments can help quantify phenotypic differences with statistical rigor .
Using ylmG as a model system for membrane protein biogenesis offers several advantages and approaches:
Investigation of membrane insertion pathways:
Create translational fusions with insertion intermediates
Develop real-time fluorescence assays for membrane integration
Identify interacting components of insertion machinery
Compare SRP-dependent and SRP-independent routing
Topology determination model:
Establish systematic approaches for topology mapping
Evaluate the roles of positive-inside rule and hydrophobicity
Test effects of sequence modifications on orientation
Examine charge distribution effects on transmembrane segments
Quality control mechanisms:
Study degradation pathways for misfolded variants
Identify chaperone interactions during membrane integration
Investigate conditional stability under stress conditions
Map quality control checkpoints in the secretion pathway
Methodological development:
Optimize detergent/lipid systems for membrane protein studies
Develop high-throughput screens for membrane protein expression
Create reporter systems for proper membrane insertion
Establish in vitro translation-translocation assays
B. subtilis represents a powerful bacterial host for academic research and industrial purposes , making it an excellent model system for studying fundamental aspects of membrane protein biogenesis in Gram-positive bacteria.
Investigating ylmG's potential role in stress response or membrane organization requires multifaceted approaches:
Stress response analysis:
Membrane organization assessment:
Lipid domain visualization using specific probes
Membrane fluidity measurements (FRAP, anisotropy)
Quantitative analysis of protein diffusion kinetics
Detergent resistance membrane isolation
Interaction with stress response machinery:
Bacterial two-hybrid screening with stress response proteins
Co-immunoprecipitation followed by MS analysis
FRET/BRET analysis of protein-protein proximity
Suppressor screening of stress response mutant phenotypes
Functional perturbation studies:
Stress response inhibition effects
Membrane stress challenges
Depletion and overexpression phenotypes
Temperature-sensitive alleles for rapid inactivation
Investigation framework table:
Hypothesis | Primary approach | Secondary validation | Expected phenotypes if true |
---|---|---|---|
Stress response component | Phenotypic analysis under stress | Gene expression analysis | Stress sensitivity in mutants |
Lipid domain organization | Flotillin co-localization | Lipid composition analysis | Altered membrane fluidity, domain disruption |
Membrane integrity factor | Membrane permeability assays | Microscopic analysis of cell morphology | Increased membrane permeability |
Protein quality control | Interaction with membrane chaperones | Misfolded protein accumulation in mutants | Protein aggregation, growth defects |
Statistical analysis of ylmG membrane localization requires specialized approaches:
Spatial statistics for localization patterns:
Ripley's K-function for clustering analysis
Pair correlation functions for spatial relationships
Nearest neighbor distance distribution
DBSCAN for density-based cluster identification
Colocalization statistics:
Pearson's correlation coefficient
Manders' overlap coefficient
Costes randomization for significance testing
Object-based colocalization analysis
Dynamic distribution analysis:
Mean square displacement analysis
Jump-distance analysis for diffusion modes
Hidden Markov Models for state transitions
Density-based trajectory classification
Statistical hypothesis testing:
Bootstrap resampling for confidence intervals
Monte Carlo simulations for pattern significance
Bayesian hierarchical modeling for complex patterns
Multiple testing correction for genome-wide screens
The statistical approach should follow principles of experimental design for big data analysis, employing appropriate sampling strategies and accounting for data heterogeneity .
Integrating diverse experimental data to build a coherent model of ylmG function requires a systematic framework:
Data standardization and quality assessment:
Normalize data across different experimental platforms
Implement quality control metrics for each data type
Assess reproducibility within and between experiments
Weight data based on methodological robustness
Multi-omics data integration:
Correlation analysis between transcriptomic and proteomic data
Network inference from protein-protein interaction studies
Pathway enrichment analysis across multiple datasets
Integrative clustering to identify functional modules
Causal relationship modeling:
Bayesian network analysis to infer directional relationships
Intervention-based experiments to validate causal models
Time-series analysis to establish temporal sequences
Perturbation response profiling
Model validation strategies:
Cross-validation across independent datasets
Prospective validation of model predictions
Sensitivity analysis to identify critical parameters
Comparison with established bacterial protein models
This integrative approach applies principles from experimental design theory and can incorporate methodologies used in studying other bacterial membrane proteins , resulting in a more robust and comprehensive functional model.