KEGG: efa:EF1580
STRING: 226185.EF1580
EF_1580 is classified as a hypothetical protein belonging to the UPF0291 protein family. Transcriptomic analyses have identified EF_1580 as one of the down-regulated genes during mammalian infection, suggesting a potential role in bacterial adaptation to changing environments . The gene appears in antisense transcript lists from RIVET (Recombination-based In Vivo Expression Technology) screens, indicating potential regulation by antisense RNA mechanisms . The genomic neighborhood analysis suggests potential co-regulation with genes involved in stress response pathways, although this requires further experimental validation.
Transcriptomic data indicates that EF_1580 expression is significantly down-regulated during growth in mammalian host environments, particularly at 8 hours post-infection . This down-regulation pattern aligns with other genes that are regulated in response to changing nutrient availability, suggesting EF_1580 may be part of the adaptive response to host environments. Comprehensive expression profiling under various conditions (pH variations, nutrient limitations, temperature shifts, and antibiotic exposures) would provide valuable insights into the regulatory network controlling EF_1580 expression.
While specific conservation data for EF_1580 is not directly provided in the search results, the approach to answering this question would involve:
Performing multiple sequence alignments of EF_1580 homologs across various enterococcal species and strains
Calculating sequence identity and similarity percentages
Identifying conserved domains and motifs
Constructing phylogenetic trees to visualize evolutionary relationships
The UPF0291 protein family is found across multiple bacterial species, suggesting potential functional importance. Conservation analysis would help establish whether EF_1580 represents a core gene within the enterococcal genome or shows strain-specific variations that might correlate with pathogenicity or niche adaptation.
The recombinant expression of EF_1580 requires careful consideration of several factors:
Expression System Selection:
E. coli BL21(DE3): Suitable for initial expression attempts due to well-established protocols
E. coli Rosetta: Preferred if EF_1580 contains rare codons
Homologous expression in E. faecalis: Valuable for maintaining native post-translational modifications
Vector Design Considerations:
Inclusion of appropriate affinity tags (His, GST, MBP) for purification
Incorporation of protease cleavage sites for tag removal
Codon optimization based on the expression host
Expression Conditions Optimization:
Testing multiple induction temperatures (16°C, 25°C, 37°C)
Varying IPTG concentrations (0.1-1.0 mM)
Evaluating different media formulations (LB, TB, auto-induction)
Solubility Enhancement Strategies:
Co-expression with molecular chaperones
Fusion with solubility-enhancing tags
Addition of osmolytes or mild detergents to lysis buffers
Assessment of expression levels and solubility through SDS-PAGE and Western blotting would guide optimization of these parameters for maximum yield of functional protein.
A multi-step purification approach is recommended:
Initial Capture:
Immobilized metal affinity chromatography (IMAC) for His-tagged constructs
Glutathione affinity chromatography for GST-fusion proteins
Intermediate Purification:
Ion exchange chromatography based on theoretical pI of EF_1580
Tag removal using specific proteases (TEV, PreScission, thrombin)
Polishing Steps:
Size exclusion chromatography to separate monomeric from aggregated forms
Removal of endotoxins if protein is intended for immunological studies
Quality Control Assessment:
SDS-PAGE and Western blotting for purity evaluation
Dynamic light scattering for aggregation analysis
Mass spectrometry for identity confirmation
Thermal shift assays for stability assessment
Optimizing buffer conditions (pH, salt concentration, additives) at each purification stage is crucial for maintaining protein stability and preventing aggregation.
Multiple complementary approaches should be employed:
| Technique | Information Obtained | Sample Requirements | Resolution |
|---|---|---|---|
| X-ray Crystallography | High-resolution 3D structure | Diffracting crystals (5-10 mg/ml) | Potentially atomic (1-3 Å) |
| NMR Spectroscopy | Structure in solution, dynamics | 15N/13C-labeled protein (1-2 mM) | Medium (dependent on size) |
| Cryo-EM | Structure, complex assembly | 50-100 μg, >100 kDa preferred | Medium to high (2-4 Å) |
| CD Spectroscopy | Secondary structure content | 0.1-0.5 mg/ml | Low (secondary structure elements) |
| SAXS | Low-resolution envelope, oligomeric state | 1-5 mg/ml | Low (10-30 Å) |
| HDX-MS | Conformational dynamics, binding interfaces | 10-100 μM | Medium (peptide level) |
For UPF0291 family proteins like EF_1580, integrating computational modeling with experimental data can provide valuable structural insights, especially when high-resolution experimental structures prove challenging to obtain.
The transcriptomic data showing down-regulation of EF_1580 during infection suggests several hypotheses regarding its role in virulence :
Metabolic Adaptation Hypothesis: EF_1580 may function in metabolic pathways active during environmental growth but down-regulated during host adaptation.
Immune Evasion Strategy: Down-regulation could represent a mechanism to alter surface properties or antigenic profiles during infection, potentially evading host immune recognition.
Stress Response Coordination: EF_1580 might participate in stress response pathways that are differentially regulated during infection.
Regulatory Role in Virulence Gene Expression: As a potential antisense regulatory target, EF_1580 could influence expression of virulence factors through RNA-based mechanisms.
Experimental approaches to investigate these hypotheses include:
Generation of EF_1580 deletion and overexpression mutants
Virulence assessment in infection models, including biofilm formation capacity
Transcriptomic and proteomic comparison of wildtype and mutant strains
Identification of interaction partners through pull-down assays and interactome studies
The observation that EF_1580 appears in antisense transcript lists suggests potential involvement in regulatory networks controlling virulence gene expression .
Given that E. faecalis is known for its ability to form biofilms and demonstrate antibiotic resistance , EF_1580's potential role in these processes merits investigation:
Biofilm Context: E. faecalis isolates from clinical and oral sources demonstrate enhanced biofilm formation capabilities compared to food isolates . If EF_1580 is involved in biofilm formation, its expression might correlate with biofilm development stages.
Experimental Approach for Biofilm Studies:
Quantitative biofilm assays comparing wildtype and EF_1580 mutants
Confocal microscopy analysis of biofilm architecture
Transcriptomic analysis during biofilm development
Assessment of cell surface properties and extracellular matrix composition
Antibiotic Resistance Connection:
Minimum inhibitory concentration (MIC) determination for various antibiotics
Stress response profiling in presence of sub-inhibitory antibiotic concentrations
Assessment of persister cell formation in EF_1580 mutants
The observation that clinical and plaque/saliva isolates show similar antibiotic resistance patterns suggests shared adaptative mechanisms that might involve proteins like EF_1580.
The RIVET antisense screen identified EF_1580 as potentially regulated by antisense transcripts during infection , suggesting a complex regulatory mechanism:
Antisense Regulation Mechanisms:
Transcriptional interference through RNA polymerase collision
Translational inhibition by blocking ribosome binding
Double-stranded RNA formation triggering RNase III degradation
Alteration of mRNA secondary structure affecting stability
Experimental Validation Approaches:
Northern blot analysis to confirm antisense transcript expression
RT-qPCR to quantify sense and antisense transcript levels
RNA-seq with strand-specific library preparation
Reporter gene assays to assess regulatory effects
Functional Consequences:
Temporal expression profiling during infection progression
Correlation with stress response and virulence gene expression
Identification of conditions triggering antisense regulation
Since approximately 9.3% of down-regulated genes at eight hours post-infection corresponded with antisense transcripts identified in the RIVET screen , this represents a significant regulatory mechanism potentially affecting E. faecalis adaptation to the host environment.
The available transcriptomic data provides several analytical approaches for understanding EF_1580:
Co-expression Network Analysis:
Identify genes with expression patterns similar to EF_1580
Construct co-expression networks to predict functional associations
Apply gene set enrichment analysis to identify biological processes
Comparative Transcriptomics:
Compare EF_1580 expression across different infection models
Analyze expression in various stress conditions
Examine regulatory patterns in different E. faecalis strains
Regulatory Element Identification:
Analyze promoter regions for transcription factor binding sites
Search for small RNA interaction sites
Identify potential antisense transcription start sites
Integration with Other Omics Data:
Correlate transcriptomic changes with proteomic profiles
Combine with metabolomic data to connect to metabolic pathways
Integrate with ChIP-seq data to identify regulatory interactions
The observation that EF_1580 was identified in both microarray and RIVET studies underscores its significance in E. faecalis gene regulation during infection.
Multiple computational strategies can provide functional insights:
| Approach | Tools | Expected Outcomes | Limitations |
|---|---|---|---|
| Homology Modeling | SWISS-MODEL, Phyre2, I-TASSER | 3D structural predictions | Accuracy depends on template availability |
| Domain Prediction | InterProScan, SMART, Pfam | Functional domain identification | Limited to known domain families |
| Structural Classification | CATH, SCOP | Fold assignment and functional inference | Requires structural data or models |
| Molecular Docking | AutoDock, HADDOCK | Potential ligand interactions | High false positive rate |
| Molecular Dynamics | GROMACS, AMBER | Dynamic behavior, conformational changes | Computationally intensive |
| Network Analysis | STRING, GeneMANIA | Functional associations, interaction networks | Based on existing data and predictions |
| Phylogenetic Profiling | OrthoMCL, InParanoid | Evolutionary conservation patterns | Requires diverse genome data |
For hypothetical proteins like EF_1580, combining structural predictions with evolutionary analysis often provides the most reliable functional hypotheses.
When faced with contradictory results:
Methodological Reconciliation:
Carefully examine experimental conditions across studies
Consider strain differences and genetic backgrounds
Evaluate methodology variations and their potential impact
Assess statistical approaches and significance thresholds
Biological Explanation Exploration:
Consider context-dependent functions
Evaluate potential pleiotropic effects
Explore post-translational modifications
Examine protein interaction networks in different conditions
Validation Strategies:
Perform independent replication studies
Use complementary methodological approaches
Develop in vitro systems that recapitulate in vivo conditions
Employ genetic approaches (knockouts, complementation, point mutations)
Integration Framework:
Develop testable models that accommodate apparently conflicting results
Design critical experiments to distinguish between alternative hypotheses
Consider mathematical modeling to integrate diverse datasets
The observation that different experimental approaches (microarray vs. RIVET) identified distinct but complementary sets of genes highlights the importance of methodological diversity in building a comprehensive understanding of bacterial gene function.
Several strategic research directions could advance understanding of EF_1580:
Genetic Manipulation Strategies:
CRISPR-Cas9 genome editing for precise genetic modifications
Conditional expression systems to study essential functions
Single-cell analysis to examine population heterogeneity
In Vivo Infection Models:
Comparison across multiple infection models (abscess, endocarditis, UTI)
Host-specific adaptation patterns
In vivo competition assays between wildtype and mutant strains
Host-Pathogen Interaction Studies:
Identification of host cell targets or receptors
Immune response profiling
Visualization of EF_1580 localization during infection
Multi-Species Interactions:
Role in polymicrobial infections
Contribution to competitive fitness against other microbes
Function in mixed biofilm communities
The observation that E. faecalis can persist in specific host niches like kidneys while also being found in diverse environments including food and oral sites suggests adaptative mechanisms potentially involving proteins like EF_1580.
Cutting-edge structural approaches offer new opportunities:
Integrative Structural Biology:
Combining multiple data sources (X-ray, NMR, cryo-EM, SAXS)
Hybrid modeling approaches
In-cell structural determination
Dynamic Structural Studies:
Time-resolved crystallography
Single-molecule FRET
Nuclear magnetic resonance relaxation dispersion
Hydrogen-deuterium exchange mass spectrometry
Structure-Guided Functional Analysis:
Alanine scanning mutagenesis of predicted functional sites
Chimeric protein construction
Structure-based inhibitor design
Computational simulations of conformational changes
Protein Interaction Mapping:
Structural characterization of protein complexes
Identification of interaction interfaces
Allosteric regulation mechanisms
Advanced structural information would complement the transcriptomic data by providing molecular-level insights into how EF_1580 functions within the cell.
Evaluating EF_1580's potential clinical applications requires:
Target Validation Studies:
Essentiality assessment in different infection models
Virulence contribution quantification
Conservation analysis across clinical isolates
Absence of human homologs
Drug Discovery Approaches:
High-throughput screening for inhibitors
Fragment-based drug design
Structure-based virtual screening
Peptide inhibitor development
Biomarker Development:
Detection of EF_1580 or antibodies against it in clinical samples
Correlation with infection severity or antibiotic resistance
Development of rapid diagnostic tests
Resistance Development Assessment:
Frequency of resistance emergence
Fitness cost of resistance mutations
Alternative pathways that might circumvent inhibition
Given E. faecalis's significant role in nosocomial infections and its concerning antibiotic resistance profiles , novel therapeutic targets like EF_1580 merit thorough investigation, especially if they contribute to virulence or antibiotic resistance mechanisms.