Recombinant E. coli uncharacterized protein yeiS refers to the engineered expression of the yeiS gene product in E. coli strains. The native yeiS gene (locus b2145/JW5359) encodes a small, uncharacterized protein of 79 amino acids (AA) with a molecular weight of approximately 9,264 Da . Key structural features include:
Domain Architecture: Belongs to the DUF2542 family, a domain of unknown function (DUF) associated with regulatory or sensory roles .
Sequence: MDVQQFFVVAVFFLIPIFCFREAWKGWRAGAIDKRVKNAPEPVYVWRAKNPGLFFAYMVA YIGFGILSIGMIVYLIFYR .
Tag: Typically expressed with an N-terminal His-tag for purification .
yeiS is hypothesized to participate in regulatory or metabolic pathways based on its predicted interactions with other E. coli proteins (Table 2). These associations were identified via computational models and co-occurrence analyses .
Interacting Protein | Proposed Role | Interaction Score |
---|---|---|
sanA | Vancomycin resistance, cell envelope integrity | 0.980 |
yfcC | Glyoxylate shunt modulation | 0.817 |
cdd | Cytidine deaminase (nucleotide metabolism) | 0.736 |
yedW | Response regulator (HprR/HprS system; H₂O₂ response) | 0.642 |
While yeiS is small and expressed in E. coli, challenges in its production and functional characterization include:
Low Functional Annotation: Despite its conservation across E. coli strains, yeiS lacks direct biochemical or phenotypic data .
Expression Optimization: General E. coli challenges (e.g., inclusion body formation, codon bias) may apply, though specific data for yeiS is absent .
Domain Complexity: The DUF2542 domain’s unknown function complicates targeted assays .
Based on interaction networks and domain predictions, yeiS may:
Regulate Stress Responses: Interactions with yedW (a two-component response regulator) imply involvement in hydrogen peroxide sensing or adaptive pathways .
Modulate Metabolic Pathways: Links to yfcC (glyoxylate shunt) and cdd (nucleotide metabolism) suggest roles in energy or nucleotide homeostasis .
Contribute to Membrane Integrity: Association with sanA (vancomycin resistance) hints at membrane-associated functions .
Functional Knockout Studies: Deletion mutants are needed to assess phenotypic impacts.
Biochemical Assays: Enzymatic activity (e.g., kinase, phosphatase) or DNA-binding capabilities require testing.
Structural Analysis: X-ray crystallography or NMR could resolve domain-specific functions.
KEGG: ecj:JW5359
STRING: 316385.ECDH10B_2302
YeiS is currently classified as an uncharacterized protein in Escherichia coli. Based on structural prediction models and comparative analysis with similar bacterial proteins, YeiS likely contains specific domain architectures that could indicate its functional role. Similar uncharacterized E. coli proteins, such as YjeQ, have been found to contain specific domain architectures including an N-terminal OB-fold RNA-binding domain, a central GTPase module, and a zinc knuckle-like C-terminal cysteine cluster . Structural analysis through methods like X-ray crystallography, NMR spectroscopy, or cryo-EM would be necessary to definitively determine YeiS structure. Functional prediction may involve analysis of conserved domains, motifs, and sequence homology with characterized proteins across bacterial species.
For recombinant expression of E. coli proteins like YeiS, several expression systems can be employed with varying efficiency. The most common approach utilizes E. coli-based expression systems with vectors containing inducible promoters (T7, lac, or tac). For optimal expression, a screening of multiple vectors with different promoters, signal peptides, and fusion tags is recommended . Expression conditions including temperature (typically testing 16°C, 25°C, 30°C, and 37°C), induction timing, and inducer concentration should be systematically evaluated. For difficult-to-express proteins, specialized E. coli strains like BL21(DE3), Rosetta, or C41/C43 may improve soluble protein yield. High-throughput screening in multiwell plate formats can efficiently identify optimal expression conditions before scaling up to shake flask or bioreactor production .
Purification of recombinant YeiS typically begins with the addition of affinity tags during cloning (His-tag being the most common) . The recommended purification workflow includes:
Cell lysis using sonication, French press, or enzymatic methods in appropriate buffer systems (typically phosphate or Tris-based buffers with protease inhibitors)
Initial capture using affinity chromatography (Ni-NTA for His-tagged proteins)
Intermediate purification via ion exchange chromatography
Polishing step using size exclusion chromatography
For analytical assessment of purity, SDS-PAGE with Coomassie or silver staining should show >90% purity . Western blotting using anti-His antibodies can confirm identity. Mass spectrometry analysis provides definitive confirmation of protein identity and integrity. Buffer optimization during purification should be conducted to maintain protein stability and prevent aggregation.
Determining the cellular localization of YeiS requires a multi-method approach:
Computational prediction: Use algorithms like PSORT, SignalP, or TMHMM to predict potential localization based on sequence features.
Fluorescent protein fusion: Generate C-terminal or N-terminal GFP/mCherry fusions of YeiS and visualize using confocal microscopy.
Cell fractionation: Separate E. coli cellular components (cytoplasm, inner membrane, periplasm, outer membrane) through sequential centrifugation and analyze fractions by Western blotting.
Immunogold electron microscopy: Using antibodies against YeiS or its affinity tag for precise subcellular localization.
Similar periplasmic proteins in E. coli, like YgiS, have been identified as components of transport systems . To confirm periplasmic localization, osmotic shock procedures followed by protein extraction and quantification from different cellular compartments would provide definitive evidence. Controls should include known cytoplasmic, periplasmic, and membrane proteins to validate fractionation quality.
To identify potential interaction partners of YeiS, researchers should employ complementary approaches:
Method | Description | Advantages | Limitations |
---|---|---|---|
Bacterial two-hybrid | Screen for protein-protein interactions in vivo | Detects interactions in bacterial environment | May miss transient interactions |
Pull-down assays | Immobilize purified YeiS and capture binding partners | Direct biochemical evidence | May lose weak interactions during washing |
Co-immunoprecipitation | Use antibodies to precipitate YeiS with interacting partners | Preserves native complexes | Requires specific antibodies |
Cross-linking MS | Covalently link interacting proteins before analysis | Captures transient interactions | Complex data analysis |
Proximity labeling | Fuse YeiS to enzymes like BioID to label nearby proteins | Maps spatial proteomics | May label proximal but non-interacting proteins |
Data analysis should include appropriate statistical methods to distinguish specific interactions from background. Validation of identified interactions should be performed using methods like surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) to determine binding parameters including affinity constants and stoichiometry.
Assessment of yeiS gene knockout effects requires a systematic approach:
Gene knockout generation: Use CRISPR-Cas9, lambda Red recombineering, or transposon mutagenesis to create ΔyeiS mutants.
Growth characterization: Compare growth curves under various conditions (different temperatures, pH values, nutrient limitations, stress conditions) between wild-type and ΔyeiS strains.
Phenotypic microarrays: Screen for metabolic differences using Biolog plates to assess carbon source utilization, chemical sensitivity, and other phenotypes.
Transcriptomic analysis: Perform RNA-Seq to identify genes differentially expressed in the knockout strain compared to wild-type.
Metabolomic profiling: Analyze changes in metabolite profiles using LC-MS or GC-MS.
For complementation studies, reintroducing the yeiS gene on a plasmid should restore the wild-type phenotype, confirming that observed effects are specifically due to yeiS deletion rather than polar effects or secondary mutations. Study design should include biological replicates (n≥3) and appropriate statistical analysis (e.g., ANOVA with post-hoc tests) to determine significance of observed differences.
High-throughput screening for optimal YeiS expression and solubility can be implemented through:
Multiwell plate-based expression: Using 96-well or 24-well formats with varying conditions including:
Automated sampling and analysis: Integration of robotic liquid handling systems with plate readers for OD600 measurements to monitor growth and protein expression levels .
Fluorescence-based solubility reporters: Fusion of YeiS with GFP variants, where fluorescence intensity correlates with proper folding and solubility.
Split-GFP complementation assays: For rapid visualization of soluble protein expression without purification.
When scaling up from screening conditions, researchers should be aware that small-scale expression results may not always directly translate to larger production scales due to differences in oxygen transfer, pH maintenance, and nutrient availability . Thus, mid-scale validation (50-100 mL cultures) is recommended before proceeding to production scale. Data analysis should incorporate both expression level and solubility percentage to identify truly optimal conditions.
Determining the 3D structure of YeiS requires a multi-technique approach:
X-ray crystallography:
Requires high-purity (>95%), homogeneous protein
Systematic screening of crystallization conditions (pH, salt, precipitants)
Co-crystallization with potential ligands or binding partners
Data collection at synchrotron facilities for high-resolution structures
Cryo-electron microscopy:
Particularly valuable for proteins resistant to crystallization
Sample vitrification optimization to prevent ice formation
Requires advanced image processing for high-resolution reconstruction
Nuclear Magnetic Resonance (NMR):
Suitable for smaller domains (<30 kDa)
Requires isotope labeling (15N, 13C) in minimal media
Provides dynamic information not available from static techniques
Integrative structural biology:
Combining low-resolution techniques (SAXS, SANS) with computational modeling
Homology modeling based on related proteins with known structures
Molecular dynamics simulations to understand conformational flexibility
For an uncharacterized protein like YeiS, initial domain prediction and construct optimization are crucial first steps before structural studies. Limited proteolysis experiments can identify stable domains suitable for structural analysis. If similar to YjeQ, which has a unique domain architecture , careful construct design that preserves domain integrity would be essential.
Analysis of potential enzymatic activity of YeiS should begin with bioinformatic predictions and follow a systematic experimental workflow:
Sequence analysis: Search for conserved catalytic motifs and active site residues through multiple sequence alignments with characterized enzymes.
Activity prediction: Based on related proteins (if YeiS contains domains similar to YjeQ's GTPase domain ), design assays to test predicted activities.
Substrate screening:
For potential GTPase/ATPase activity: Measure phosphate release using malachite green assay
For hydrolytic enzymes: Use fluorogenic or chromogenic substrates
For transferases: Employ radiolabeled substrates or coupled enzyme assays
Enzyme kinetics determination:
Measure initial rates at varying substrate concentrations
Determine kinetic parameters (kcat, Km, kcat/Km)
Test effects of pH, temperature, and buffer components on activity
Assess potential allosteric regulators or inhibitors
If YeiS shows enzymatic activity similar to YjeQ, both steady state and pre-steady state kinetics should be measured . For steady state measurements, spectrophotometric methods can continuously monitor product formation or substrate depletion. Pre-steady state kinetics using rapid kinetic techniques (stopped-flow or quench-flow) can reveal transient intermediates and rate-limiting steps in the catalytic cycle.
Determining the physiological role of an uncharacterized protein like YeiS requires an integrated approach:
Comparative genomics:
Analyze gene neighborhood conservation across bacterial species
Identify co-occurrence patterns with genes of known function
Examine evolutionary conservation to assess functional importance
Transcriptomic profiling:
RNA-Seq analysis under various growth conditions to identify conditions where yeiS is differentially expressed
Single-cell RNA-Seq to detect potential heterogeneity in expression
ChIP-Seq to identify transcription factors regulating yeiS expression
Growth phenotyping:
Compare wild-type and ΔyeiS strains under various stress conditions (temperature, pH, nutrient limitation, antibiotics)
High-throughput phenotypic microarrays to identify specific growth conditions affected by yeiS deletion
Competition assays to assess fitness effects
Suppressor mutation analysis:
Screen for mutations that suppress phenotypes of yeiS deletion
Whole genome sequencing of suppressor strains to identify compensatory pathways
If YeiS functions similarly to other characterized E. coli proteins, such as the deoxycholate-binding periplasmic protein YgiS , specific assays to test transport functions would be appropriate, including monitoring uptake or export of predicted substrates using radioactive or fluorescently labeled compounds.
To investigate YeiS involvement in stress response:
Expression analysis during stress conditions:
qRT-PCR or reporter gene fusions to monitor yeiS expression under various stresses (oxidative, acid, osmotic, antibiotic)
Western blotting to quantify YeiS protein levels during stress response
Promoter analysis to identify stress-responsive regulatory elements
Stress sensitivity phenotyping:
Compare survival rates of wild-type and ΔyeiS strains exposed to stress conditions
Measure growth inhibition zones in disc diffusion assays with various stressors
Time-kill curves during stress exposure
Protein interaction studies under stress:
Pull-down assays or co-immunoprecipitation under normal and stress conditions
Interactome changes using BioID or APEX proximity labeling during stress
Protein localization changes during stress using fluorescent protein fusions
Multi-omics integration:
Combine transcriptomics, proteomics, and metabolomics data to place YeiS in stress response networks
Network analysis to identify functional modules where YeiS operates
The experimental design should include appropriate positive controls, such as known stress response genes, and negative controls. If YeiS is involved in bile acid resistance similarly to YgiS , specific assays testing growth in the presence of bile acids or deoxycholate would be particularly informative.
To investigate YeiS's potential role in pathogenesis:
Virulence model systems:
Cell culture infection models comparing wild-type and ΔyeiS strains
Invasion and adhesion assays with epithelial cell lines
Survival within macrophages or other immune cells
Animal infection models with appropriate ethical approval
Host response analysis:
Cytokine production measurement following infection
Transcriptomics of host cells infected with wild-type vs. ΔyeiS strains
Inflammasome activation and pyroptosis assessment
Virulence factor expression and secretion:
Analysis of secretome differences between wild-type and ΔyeiS strains
Effects on type III secretion system functioning
Biofilm formation capacity assessment
In vivo expression and importance:
In vivo expression technology (IVET) to determine if yeiS is upregulated during infection
Transposon sequencing (Tn-Seq) to assess fitness contribution during infection
Competitive index determination in mixed infections
If YeiS functions similarly to YgiS in deoxycholate binding , its role in bile resistance during intestinal colonization would be particularly relevant to investigate. Bile acids are important host defense molecules in the gastrointestinal tract, and bacterial mechanisms to resist their effects can be critical virulence determinants.
Developing effective site-directed mutagenesis strategies requires:
Target residue identification:
Sequence conservation analysis across homologs to identify evolutionarily conserved residues
Structural modeling to predict functionally important regions
Alignment with characterized proteins to identify potential catalytic or binding residues
Mutagenesis approach selection:
QuikChange PCR for single mutations
Gibson Assembly for multiple mutations or challenging regions
Golden Gate Assembly for systematic mutation libraries
Rational mutation design:
Conservative mutations (similar properties) to test structural importance
Non-conservative mutations to disrupt function
Alanine scanning of predicted functional regions
Cysteine scanning for accessibility and crosslinking studies
Functional validation pipeline:
Expression and solubility testing of mutants
Biochemical assays to measure altered activity
Binding studies with potential interaction partners
In vivo complementation tests in ΔyeiS strains
For proteins with GTPase activity similar to YjeQ , mutations in the G1 motif (such as S-to-A mutations in the P-loop) would be particularly informative to test nucleotide binding and hydrolysis functions. A comprehensive mutagenesis approach would include mutations in predicted catalytic residues, substrate-binding regions, and domain interface residues to understand both function and interdomain communication.
To study yeiS gene expression regulation effectively:
Promoter characterization:
5' RACE to precisely map transcription start sites
Reporter gene fusions (lacZ, gfp) with promoter fragments of various lengths
ChIP-seq to identify transcription factor binding sites
In vitro DNA-protein interaction studies (EMSA, DNase footprinting)
Transcriptional regulation analysis:
RNA-Seq under various conditions to identify expression patterns
Quantitative RT-PCR to validate specific expression changes
Transcription factor overexpression/deletion effects on yeiS expression
Global transcription machinery engineering to identify regulatory dependencies
Post-transcriptional regulation:
mRNA stability measurements through rifampicin chase experiments
Identification of potential regulatory small RNAs through co-expression analysis
RNA structure probing to identify regulatory elements
Translational efficiency assessment using ribosome profiling
Environmental response characterization:
Systematic testing of expression under different conditions (nutrients, temperature, pH, osmolarity)
Correlation of expression with specific cellular processes or stress responses
Particular attention should be paid to conditions where related proteins like YgiS or YjeQ show altered expression. The presence of deoxycholate, which affects YgiS expression , might be a relevant condition to test for yeiS regulation as well. Integration of expression data with information about cellular processes affected in ΔyeiS strains can provide insights into the physiological context of gene regulation.
Computational prediction of YeiS functional networks should employ multiple complementary approaches:
Sequence-based methods:
Phylogenetic profiling to identify genes with similar evolutionary patterns
Gene neighborhood analysis to find consistently co-located genes
Domain fusion analysis to detect functional relationships
Protein-protein interaction prediction based on sequence features
Structure-based approaches:
Structural similarity searches to identify proteins with similar folds
Protein-protein docking with predicted interaction partners
Molecular dynamics simulations to assess dynamic behavior
Binding site prediction and comparison with characterized proteins
Network inference methods:
Co-expression network analysis across multiple datasets
Bayesian network inference from multi-omics data
Text mining of scientific literature for functional associations
Metabolic network modeling to predict pathway involvement
Integrative approaches:
Weighted integration of multiple evidence types for function prediction
Machine learning models trained on known functional relationships
Network clustering to identify functional modules containing YeiS
For validation, predictions should be tested experimentally through targeted approaches such as co-immunoprecipitation, bacterial two-hybrid, or phenotypic analysis of double mutants. If YeiS functions in transport systems similar to YgiS , particular attention should be paid to predicting potential transported substrates and associated components of the transport machinery.