Listeria monocytogenes is a Gram-positive, rod-shaped bacterium known for its ability to survive in diverse and extreme conditions, including a wide range of temperatures, pH levels, and salt concentrations . As a facultative anaerobe, L. monocytogenes can grow with or without oxygen, and it does not form spores . This bacterium is a significant foodborne pathogen that can cause listeriosis, a severe infection resulting from the consumption of contaminated food . L. monocytogenes has the capability to invade and reproduce inside host cells, making it particularly virulent .
L. monocytogenes is classified into different serotypes, with serotype 4b being frequently associated with epidemic human listeriosis outbreaks . Genetic analysis has identified three main lineages within L. monocytogenes:
Serotypes in lineage I are commonly linked to human listeriosis, while lineage II also contains serotypes associated with human disease, and lineage III has fewer reported isolates .
Translation initiation factor IF-2 (infB) is a crucial protein involved in the initiation of protein synthesis in bacteria. The infB gene encodes this factor, which plays a vital role in the accurate and efficient translation of mRNA into proteins.
Internalin F (inlF) is a surface protein that contributes to the virulence of L. monocytogenes . Although inlF was identified over 20 years ago, its specific role in the anti-inflammatory response of L. monocytogenes remained unclear until more recent studies . Research indicates that inlF is highly expressed during the infection of non-phagocytic cells and contributes to the adhesion and invasion of macrophages . Deletion of inlF has been shown to reduce the expression of pro-inflammatory cytokines, such as interleukin (IL)-1β and tumor necrosis factor (TNF-α), suggesting its role in modulating the host's immune response . In a study using mice, inlF significantly inhibited the expression of IL-1β and IL-6 in the spleen and IL-1β, IL-6, and TNF-α in the liver . Furthermore, inlF contributes to the colonization of L. monocytogenes in the spleen, liver, and ileum during the early stages of infection, leading to severe inflammatory injury and histopathologic changes in the later stages .
Genetic analysis has revealed that certain strains of serotype 4b exhibit divergence in specific regions . These divergent regions are of particular interest because they may contribute to the unique characteristics and virulence of these strains . Identifying these genetic markers can aid in the detection and characterization of strains belonging to specific lineages .
Interferons (IFNs) are a family of cytokines that play a crucial role in both innate and adaptive immune responses to infections . While IFNs are well-known for their antiviral activities, their role in bacterial infections is more complex . Different strains of L. monocytogenes vary significantly in their ability to induce IFN-β, with some strains, like LO28, hyperinducing IFN-β due to a non-functional BrtA .
L. monocytogenes interacts with host cells through surface proteins like internalins. These interactions facilitate the bacterium's entry into non-phagocytic cells and macrophages . Studies have shown that inlF is significantly upregulated during infection of various cell types, suggesting its importance in the interaction between L. monocytogenes and host cells .
KEGG: lmf:LMOf2365_1342
Translation Initiation Factor IF-2 (infB) plays a critical role in bacterial protein synthesis by facilitating the binding of initiator tRNA (fMet-tRNA) to the 30S ribosomal subunit during translation initiation. In Listeria monocytogenes serotype 4b, IF-2 functions similarly to other bacterial species, promoting proper formation of the initiation complex and ensuring accurate translation of mRNA . This protein is essential for bacterial survival and virulence expression, as it enables the pathogen to synthesize proteins necessary for host invasion and immune evasion. Serotype 4b strains, particularly the 4bV variant, contain distinctive genomic elements that may influence protein expression patterns, including translation initiation factors .
Listeria monocytogenes serotype 4b is distinguished by its significantly higher association with listeriosis outbreaks compared to other serotypes. Recent research has identified variant strains (4bV) that contain a 6.3 kb segment of DNA normally restricted to lineage II strains while maintaining the serological characteristics of 4b strains . This genetic region contains six genes (lmo0734–lmo0739) that encode proteins with various functions, including a transcriptional regulator, metabolic enzymes, and transport systems . The 4bV strains have been linked to multiple listeriosis outbreaks in 2014-2016 in the USA, highlighting their clinical importance. Unlike other serotypes, serotype 4b (along with 1/2a and 1/2b) is responsible for the majority of human listeriosis cases, making its molecular components, including translation factors, particularly relevant for clinical research .
Recombinant expression of Listeria monocytogenes proteins, including translation factors like IF-2, typically employs Escherichia coli expression systems. Based on established protocols for similar Listeria proteins, the process involves:
Gene amplification: PCR amplification of the infB gene from L. monocytogenes serotype 4b genomic DNA
Vector construction: Cloning into expression vectors containing appropriate promoters and affinity tags
Expression conditions: Optimization of temperature, IPTG concentration, and induction time for maximal protein yield
Purification: Affinity chromatography using His-tag or other fusion tags, followed by size exclusion chromatography
For internalin proteins from L. monocytogenes, researchers have successfully used pAE vectors for expression in E. coli, followed by protein A-affinity chromatography for purification . Similar approaches can be adapted for the expression of translation initiation factors, with modifications to account for protein-specific characteristics.
Integrating whole genome sequencing with recombinant protein studies provides a powerful approach to characterizing IF-2 variants in L. monocytogenes serotype 4b strains. A comprehensive methodological framework includes:
Genomic analysis: Use WGS data to identify polymorphisms in the infB gene across different L. monocytogenes 4b isolates. Apply the CFSAN SNP Pipeline to detect single nucleotide polymorphisms that might affect protein function .
Variant selection: Prioritize IF-2 variants based on:
Proximity to functional domains
Conservation across strains
Association with virulence or antibiotic resistance
Recombinant expression: Express identified variants as recombinant proteins using optimized E. coli systems.
Functional characterization: Compare biochemical properties of wild-type and variant IF-2 proteins through:
Ribosome binding assays
GTPase activity measurements
Thermal stability analyses
Structural studies: Determine if amino acid substitutions alter protein structure using X-ray crystallography or cryo-EM.
This integrated approach has successfully revealed important insights into other L. monocytogenes proteins and could be particularly valuable for understanding the functional significance of conservation or variation in translation factors across different serotype 4b strains, including the clinically important 4bV variants .
Developing recombinant protein-based detection methods for 4bV strains presents several significant challenges:
Serological similarity: 4bV strains are serologically identical to 4b strains despite containing a 6.3 kb segment of DNA normally restricted to lineage II strains, making antibody-based discrimination difficult .
Conserved epitopes: Traditional antibodies might not distinguish between standard 4b and 4bV strains if targeting highly conserved epitopes.
Technical solutions:
Generate antibodies against proteins encoded by the lineage II-specific genes (lmo0734–lmo0739) present in 4bV strains
Design sandwich ELISA systems using combinations of antibodies against both common and variant-specific targets
Develop phage-display antibody libraries against specific epitopes unique to 4bV strains
Validation requirements: Any new detection method requires extensive validation against diverse strain collections, including:
| Strain Type | Number of Isolates | Geographical Origins | Source Types |
|---|---|---|---|
| 4b standard | ≥30 | Multiple countries | Clinical, food, environmental |
| 4bV variants | ≥30 | USA, Australia, Chile | Clinical, food, environmental |
| Other serotypes | ≥50 | Global | Various |
Recent successes with phage display-derived monoclonal antibodies against InlA and InlB suggest similar approaches might be applicable for developing detection systems that can distinguish between standard 4b and 4bV strains based on their unique protein profiles .
Optimizing translation efficiency of recombinant L. monocytogenes IF-2 in heterologous systems requires a multifaceted approach:
Codon optimization: Analyze codon usage bias between L. monocytogenes and the expression host (typically E. coli). Adjust rare codons to match the host's preferences while maintaining critical structural elements.
Vector selection: Choose expression vectors with:
Strong, inducible promoters (T7, tac)
Optimal ribosome binding sites
Appropriate fusion tags that don't interfere with IF-2 function
Expression conditions optimization:
Test multiple induction temperatures (16°C, 25°C, 30°C, 37°C)
Vary inducer concentration (0.1-1.0 mM IPTG)
Optimize growth media (rich vs. minimal, supplemented with amino acids)
Consider co-expression with chaperones for proper folding
Protein solubility enhancement:
Use solubility-enhancing tags (SUMO, MBP, TRX)
Test different cell lysis conditions to minimize aggregation
Employ additives (arginine, detergents) in purification buffers
Activity preservation:
Include GTP in buffers to stabilize IF-2
Minimize freeze-thaw cycles
Determine optimal storage conditions through activity assays
Researchers working with similar recombinant proteins from L. monocytogenes have successfully preserved activity by carefully optimizing expression conditions and purification protocols, as demonstrated with recombinant InlA and InlB proteins .
Translation initiation factor IF-2 from L. monocytogenes serotype 4b shares the core functional domains with other bacterial pathogens but exhibits several distinguishing features:
Structural comparison:
Contains conserved G-domain (GTPase) and C2 domain for ribosome binding
May possess unique linker regions that influence flexibility and interactions with Listeria-specific ribosomal components
Predicted to maintain the three-domain architecture typical of bacterial IF-2 proteins
Functional distinctions:
Adapted to function optimally at lower temperatures consistent with Listeria's psychrotrophic nature
May exhibit differential GTP hydrolysis rates compared to enteric pathogens
Potentially shows altered interactions with fMet-tRNA that reflect Listeria's adaptation to different host environments
Regulatory mechanisms:
The expression and activity of IF-2 in L. monocytogenes likely responds to environmental stresses encountered during infection
May be subject to post-translational modifications unique to Listeria's intracellular lifestyle
Research approaches comparing IF-2 across diverse bacterial species could help identify pathogen-specific characteristics that might be exploited for targeted antimicrobial development. Similar comparative approaches have been successfully applied to other Listeria proteins, revealing important insights into their functional adaptation .
Investigating the role of IF-2 in L. monocytogenes stress response and virulence requires a multi-faceted experimental approach:
Gene expression analysis:
qRT-PCR to measure infB expression under various stress conditions (acid, oxidative, heat, cold stresses)
RNA-seq to identify co-regulated genes in stress response networks
Ribosome profiling to assess translation efficiency during stress adaptation
Genetic manipulation strategies:
Construction of conditional infB mutants (since complete deletion may be lethal)
Site-directed mutagenesis of key functional domains
CRISPR interference for partial knockdown of expression
Protein-level analyses:
Pull-down assays to identify IF-2 interaction partners during infection
Phosphoproteomics to detect stress-induced post-translational modifications
In vitro translation assays comparing activity under various stress conditions
Infection models:
Tissue culture invasion and intracellular replication assays with IF-2 mutants
Murine infection models to assess virulence in vivo
Competition assays between wild-type and IF-2 variant strains
Structural biology approaches:
Cryo-EM studies of IF-2 bound to Listeria ribosomes
Hydrogen-deuterium exchange mass spectrometry to identify conformational changes upon stress
These methodological approaches build upon successful strategies used to characterize other L. monocytogenes virulence factors, such as the internalins, and can provide insights into how translation factors contribute to pathogenesis .
Recombinant IF-2 offers promising opportunities for developing novel diagnostic tools for L. monocytogenes serotype 4b detection, particularly for difficult-to-identify variant strains:
Antibody development:
Aptamer-based detection:
Select RNA or DNA aptamers with high affinity for L. monocytogenes IF-2
Incorporate aptamers into lateral flow devices or biosensors
Optimize detection limits to reach 10³ CFU/mL or better
Recombinant phage-based detection:
Multiplex approaches:
Combine IF-2 detection with other serotype-specific markers
Develop assays that simultaneously detect multiple virulence factors
Create diagnostic panels that differentiate between standard 4b and 4bV strains
| Detection Method | Sensitivity (CFU/mL) | Time to Result | Sample Compatibility |
|---|---|---|---|
| Antibody-based ELISA | 10⁴-10⁵ | 3-4 hours | Food, clinical |
| Aptamer biosensors | 10³-10⁴ | 30-60 minutes | Food, environmental |
| Phage-based detection | 10²-10³ | 1-2 hours | Food, environmental |
| Multiplex PCR | 10³-10⁴ | 2-3 hours | All sample types |
These approaches build on successful strategies for developing diagnostic tools for Listeria detection, as evidenced by recent advances in antibody development and phage-based methods .
Expressing recombinant L. monocytogenes IF-2 in E. coli often presents solubility challenges that can be addressed through multiple complementary strategies:
Fusion partners optimization:
Test multiple solubility-enhancing tags in parallel:
Maltose-binding protein (MBP)
Small ubiquitin-like modifier (SUMO)
Thioredoxin (TRX)
Glutathione S-transferase (GST)
Evaluate different tag positions (N-terminal vs. C-terminal)
Incorporate flexible linkers between tag and IF-2
Expression condition modifications:
Reduce expression temperature to 16-20°C
Decrease inducer concentration (0.1-0.2 mM IPTG)
Use specialized E. coli strains (Rosetta, Arctic Express, SHuffle)
Test auto-induction media formulations
Co-expression strategies:
Co-express with molecular chaperones (GroEL/GroES, DnaK/DnaJ)
Include rare tRNA supplementation
Co-express with interaction partners that may stabilize IF-2
Domain-based approaches:
Express individual domains separately
Design truncated constructs based on structural predictions
Create domain-swapped chimeras with well-expressing bacterial homologs
Similar approaches have proven successful for expressing other challenging Listeria proteins, including surface-associated virulence factors like InlA and InlB .
Developing IF-2-based detection methods that can reliably distinguish between pathogenic and non-pathogenic Listeria species requires careful consideration of multiple factors:
Sequence analysis approach:
Perform comprehensive alignment of infB sequences across all Listeria species
Identify regions with species-specific signatures, particularly in L. monocytogenes
Design primers or probes targeting these discriminatory regions
Epitope mapping strategy:
Use phage display to identify peptide epitopes unique to L. monocytogenes IF-2
Generate monoclonal antibodies against these specific epitopes
Validate antibody specificity against a panel of Listeria species
Multiplex detection systems:
Combine IF-2 detection with established pathogenicity markers (e.g., internalins)
Develop assays that simultaneously detect multiple virulence-associated proteins
Create diagnostic algorithms that integrate multiple biomarkers for increased specificity
Validation requirements:
| Listeria Species | Minimum Strains | Sources | Testing Parameters |
|---|---|---|---|
| L. monocytogenes | 20+ (multiple serovars) | Clinical, food, environmental | Sensitivity, specificity, limit of detection |
| L. innocua | 10+ | Food, environmental | Cross-reactivity assessment |
| Other Listeria spp. | 5+ each | Various | False positive evaluation |
| Non-Listeria controls | 10+ | Food matrices | Matrix interference |
Research has shown that combining targets like IF-2 with species-specific markers like InlA and InlB can achieve 100% sensitivity (CI 29.24–100.0) and specificity (CI 88.78–100.0) in distinguishing pathogenic from non-pathogenic Listeria, providing a model for robust diagnostic development .
Ensuring the quality and consistency of recombinant L. monocytogenes IF-2 preparations requires rigorous quality control measures across multiple parameters:
Purity assessment:
SDS-PAGE analysis (target: >95% purity)
Size exclusion chromatography to detect aggregates
Mass spectrometry for molecular weight confirmation and contaminant detection
Endotoxin testing (<1 EU/mg protein for functional studies)
Structural integrity verification:
Circular dichroism spectroscopy to confirm secondary structure
Thermal shift assays to assess stability
Limited proteolysis to verify proper folding
Dynamic light scattering to evaluate size distribution
Functional activity testing:
GTPase activity assays (comparative analysis with commercial standards)
30S ribosomal subunit binding assays
fMet-tRNA binding capacity
In vitro translation initiation efficiency
Storage stability monitoring:
Activity retention after freeze-thaw cycles
Long-term stability at -80°C, -20°C, and 4°C
Buffer optimization for maintained functionality
Aggregation assessment over time
Batch-to-batch consistency:
Standardized activity units definition
Reference standard comparison for each batch
Lot-specific certificate of analysis documentation
These quality control parameters ensure that recombinant IF-2 preparations are suitable for downstream applications in structural biology, functional characterization, and diagnostic development. Similar quality control approaches have been successfully implemented for other recombinant Listeria proteins used in diagnostic and research applications .
The epidemiological success of L. monocytogenes 4bV strains may be influenced by variations in translation factors like IF-2 through several potential mechanisms:
Translation efficiency adaptation:
IF-2 variants might optimize translation initiation under specific environmental conditions
Enhanced translation of stress response proteins could provide survival advantages
Altered translation preferences may influence the expression of virulence factors
Host interaction modulation:
Modified translational machinery could affect the production of surface proteins involved in host cell invasion
Variations in translation factors might influence the bacterial response to host immune defenses
Subtle changes in virulence factor expression timing may enhance pathogenicity
Stress response coordination:
Research approaches to test these hypotheses:
Comparative ribosome profiling between standard 4b and 4bV strains
Proteomics analysis under various stress conditions
Experimental evolution studies under selective pressures
Genetic complementation experiments swapping IF-2 variants between strains
The adaptation of translational machinery in 4bV strains represents an intriguing research direction that may explain their involvement in recent listeriosis outbreaks and their apparent emergence as significant pathogens .
Translation initiation factor IF-2 presents a promising target for novel antimicrobial development against L. monocytogenes based on several advantageous characteristics:
Target validation rationale:
IF-2 is essential for bacterial survival
The protein has sufficient structural differences from human translation factors
Translation initiation represents a bottleneck in protein synthesis
Drug discovery approaches:
High-throughput screening against the GTPase domain
Fragment-based drug design targeting IF-2/ribosome interface
Structure-based virtual screening for binding pocket inhibitors
Repurposing of existing translation inhibitors with optimization for Listeria-specificity
Potential advantages of IF-2 inhibitors:
May be effective against antibiotic-resistant L. monocytogenes strains
Could have activity against both replicating and slow-growing intracellular bacteria
Might show synergistic effects with existing antibiotics
Development challenges to address:
Ensuring selectivity over human translation factors
Achieving sufficient penetration into mammalian cells to reach intracellular bacteria
Optimizing pharmacokinetic properties for clinical applications
| Inhibitor Type | Target Site | Development Stage | Potential Advantages |
|---|---|---|---|
| GTP analogs | GTP-binding pocket | Early discovery | Direct inhibition of activity |
| Peptide mimetics | fMet-tRNA binding site | Computational design | High specificity |
| Small molecules | Allosteric sites | Virtual screening | Better drug-like properties |
| RNA aptamers | Protein-ribosome interface | Proof-of-concept | Novel mechanism of action |
This approach aligns with the growing need for new antimicrobial strategies against Listeria, particularly given increasing concerns about antibiotic resistance in clinical isolates .
Advanced structural biology approaches offer powerful tools to elucidate the precise mechanisms of L. monocytogenes IF-2 function in translation initiation:
Cryo-electron microscopy applications:
Capture IF-2 in complex with Listeria-specific ribosomes at different functional states
Resolve structures at near-atomic resolution (2-3 Å)
Visualize conformational changes during GTP hydrolysis and fMet-tRNA positioning
Compare structures between standard 4b and variant 4bV strains
X-ray crystallography strategies:
Determine high-resolution structures of individual IF-2 domains
Co-crystallize with GTP/GDP and ribosome components
Analyze binding sites for potential inhibitor development
Map serotype-specific structural variations
Integrative structural approaches:
Combine small-angle X-ray scattering (SAXS) with NMR for full-length protein dynamics
Apply hydrogen-deuterium exchange mass spectrometry to identify flexible regions
Use cross-linking mass spectrometry to map interaction interfaces
Employ molecular dynamics simulations to predict functional motions
Functional correlation analyses:
Connect structural features to biochemical activities
Identify structural determinants of temperature sensitivity
Map species-specific structural elements that could influence host adaptation
Correlate structural variations with translation efficiency differences
These structural biology approaches would provide unprecedented insights into how L. monocytogenes IF-2 functions at the molecular level, potentially revealing unique adaptations that contribute to this pathogen's virulence and environmental persistence. Similar structural approaches have been successfully applied to other bacterial translation factors, revealing important mechanistic insights into their function .