Recombinant Listeria monocytogenes serotype 4b tRNA-specific 2-thiouridylase mnmA (mnmA) is a recombinant protein derived from the bacterium Listeria monocytogenes, specifically from serotype 4b. This protein is involved in the modification of tRNA by incorporating 2-thiouridine, which is crucial for the stability and function of tRNA in bacteria. The mnmA protein is expressed in various host systems such as E. coli, yeast, baculovirus, or mammalian cells for research purposes .
Listeria monocytogenes is a Gram-positive, facultative anaerobic bacterium known for its virulence as a foodborne pathogen. Serotype 4b is particularly notable for its involvement in severe outbreaks of listeriosis, often linked to contaminated food products like unpasteurized dairy and raw foods . This serotype can infect the brain, spinal cord membranes, and bloodstream, making it highly dangerous, especially to vulnerable populations such as the elderly and immunocompromised individuals .
While the mnmA protein itself is not directly implicated in the virulence mechanisms of Listeria monocytogenes, its role in tRNA modification is essential for bacterial survival and replication. The ability of L. monocytogenes to grow and reproduce inside host cells is a key factor in its pathogenicity . Understanding the genetic and molecular components like mnmA can provide insights into how L. monocytogenes maintains its viability and infectivity.
Serotype 4b strains of L. monocytogenes exhibit distinct molecular features, including specific genetic markers that differentiate them from other serotypes. Lineages I and III of serotype 4b have been studied extensively to understand their virulence and epidemiological significance . The presence of serotype-specific genes like gtcA, gltA, and gltB is crucial for identifying and characterizing these strains .
| Characteristic | Description |
|---|---|
| Source | E. coli, Yeast, Baculovirus, Mammalian Cells |
| Target | Listeria monocytogenes Serotype 4b |
| Type | Recombinant Protein |
| Applications | Research, Potential Diagnostic/Vaccine Development |
| Purity | Variable depending on production method |
| Storage | Typically at -20°C or -80°C |
| Feature | Description |
|---|---|
| Serotype-Specific Genes | gtcA, gltA, gltB |
| Lineages | I and III |
| Virulence Factors | Beta hemolysin, ability to invade host cells |
| Pathogenicity | High replication in monocytes/macrophages |
KEGG: lmf:LMOf2365_1531
The mnmA enzyme in Listeria monocytogenes, similar to other bacterial species, is involved in the 2-thiolation (s2U) modification of the wobble uridine (U34) in tRNALys, tRNAGlu, and tRNAGln. This post-transcriptional modification is essential for maintaining tRNA structural stability, ensuring proper aminoacylation, and enhancing the precision and efficiency of codon recognition during protein translation . The process involves a "sulfur trafficking system" initiated by cysteine desulfurase and a "modification enzyme" that directly incorporates sulfur into specific tRNAs . These modifications are critical for L. monocytogenes survival and virulence by ensuring accurate protein synthesis.
While direct evidence specific to L. monocytogenes mnmA is limited, we can infer its importance based on findings from other organisms. In Toxoplasma gondii, knockout of the homologous TgMnmA gene led to significant abnormalities in apicoplast biogenesis and severely disturbed genomic transcription . By extension, mnmA likely plays a crucial role in L. monocytogenes virulence by ensuring proper protein synthesis necessary for multiple pathogenic mechanisms, including invasion of intestinal epithelial cells, intracellular replication, and evasion of host immune responses . As a facultative intracellular pathogen, L. monocytogenes relies on precise translation of virulence factors, a process potentially dependent on mnmA-mediated tRNA modifications.
The L. monocytogenes mnmA protein belongs to the tRNA-specific 2-thiouridylase family, which contains conserved domains for ATP binding and sulfur transfer activities. Similar to other bacterial mnmA proteins, it likely possesses a PP-loop domain characteristic of the adenine nucleotide alpha hydrolase superfamily that catalyzes the formation of the thiocarboxylate intermediate necessary for the 2-thiolation reaction. The enzyme requires ATP for activation of the target uridine and contains binding sites for tRNA recognition. Structural analysis using tools like HMMER would reveal specific domains and motifs similar to those found in homologous tRNA thiouridylases across bacterial species .
For optimal expression of recombinant L. monocytogenes mnmA, researchers should consider the following protocol:
Expression System Selection: E. coli BL21(DE3) strain is recommended due to its reduced protease activity and compatibility with T7 promoter-based expression vectors.
Vector Construction: Clone the L. monocytogenes mnmA gene into pET-28a(+) with an N-terminal His-tag for purification purposes.
Expression Conditions:
Culture medium: LB broth with appropriate antibiotics
Growth temperature: 37°C until OD600 reaches 0.6-0.8
Induction: 0.5-1.0 mM IPTG
Post-induction conditions: 18-20°C for 16-18 hours to maximize soluble protein yield
Purification Strategy:
Lyse cells using sonication in buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10% glycerol, and 10 mM imidazole
Purify using Ni-NTA affinity chromatography followed by size exclusion chromatography
Final storage buffer: 25 mM Tris-HCl (pH 7.5), 150 mM NaCl, 5 mM DTT, and 10% glycerol
An effective CRISPR/Cas9-based knockout strategy for mnmA in L. monocytogenes should include:
Guide RNA Design:
Identify PAM sequences (NGG) within the mnmA coding region
Design 20-nt guide sequences with minimal off-target potential
Prioritize targets in the N-terminal coding region to ensure complete loss of function
Delivery System:
Verification Methods:
PCR screening of transformants
qRT-PCR to confirm absence of mnmA transcription
Western blot analysis to confirm protein knockout
Phenotypic assays to assess functional consequences
Complementation Strategy:
To evaluate recombinant mnmA activity in vitro, researchers can employ the following methodological approaches:
Thiouridylation Assay:
Prepare in vitro transcribed tRNA substrates (tRNALys, tRNAGlu, and tRNAGln)
Incubate purified mnmA with tRNA substrates in buffer containing ATP, Mg2+, and a sulfur donor
Detect thiolated tRNAs using:
a) HPLC analysis with UV detection at 314 nm (characteristic of s2U)
b) Mass spectrometry to identify the mass shift associated with the thio modification
c) Reverse-phase thin-layer chromatography following nuclease digestion
ATP Hydrolysis Assay:
Monitor ATP consumption using a coupled enzymatic assay or 32P-labeled ATP
Compare rates with and without tRNA substrates to confirm specificity
Thermal Stability Assay:
Compare melting temperatures of native and thiolated tRNAs using UV spectroscopy
Higher melting temperatures indicate successful modification and enhanced tRNA stability
L. monocytogenes utilizes multiple redundant mechanisms for dissemination from intestinal sites to mesenteric lymph nodes (MLN) and systemic circulation . While the direct role of mnmA in this process has not been specifically characterized, the enzyme likely contributes to dissemination through:
Translation Efficiency: As a tRNA-modifying enzyme, mnmA ensures optimal translation of key virulence factors necessary for:
Stress Response Regulation: Proper tRNA modification is crucial for adaptation to stress conditions encountered during:
Contribution to Multiple Dissemination Pathways: L. monocytogenes can disseminate via:
The ability to utilize these redundant pathways may depend on proper protein synthesis facilitated by mnmA-mediated tRNA modifications, although direct experimental evidence linking mnmA to specific dissemination mechanisms remains to be established.
Environmental stress factors likely modulate mnmA expression and activity in L. monocytogenes through multiple regulatory mechanisms:
| Environmental Stress | Predicted Impact on mnmA | Physiological Consequence | Assessment Method |
|---|---|---|---|
| Acid stress (pH 5.0) | Upregulation | Enhanced translation fidelity under stress | qRT-PCR, Western blot |
| Oxidative stress (H₂O₂) | Initial downregulation followed by compensatory upregulation | Temporary translation pause followed by stress adaptation | Transcriptomics, proteomics |
| Nutrient limitation | Maintained expression | Prioritized translation of survival genes | Ribosome profiling |
| Temperature shift (4°C) | Increased activity with stable expression | Enhanced cold adaptation through codon optimization | In vitro activity assays at various temperatures |
| Bile exposure | Potential inhibition of enzymatic activity | Reduced virulence in intestinal environment | Bile sensitivity assays with mnmA mutants |
These responses likely contribute to L. monocytogenes' remarkable adaptability across diverse environments, from food preservation conditions to host physiological niches. Researchers can verify these predictions through comparative transcriptomics and proteomics of wild-type versus mnmA-deficient strains under various stress conditions.
The mnmA-mediated tRNA modification likely influences antibiotic susceptibility in L. monocytogenes through several mechanisms:
Translation Accuracy Effects:
Diminished tRNA modifications can cause translational frameshifting and misincorporation of amino acids
This may alter the production of proteins involved in antibiotic resistance, potentially:
a) Reducing expression of efflux pumps (increased susceptibility)
b) Compromising cell wall synthesis (increased susceptibility to β-lactams)
c) Altering ribosomal protein synthesis (affecting aminoglycoside targets)
Stress Response Modulation:
Proper tRNA modification by mnmA ensures efficient translation of stress response regulators
Defects may impair adaptive responses to antibiotic-induced stress
Metabolic Alterations:
Changes in translational efficiency can affect metabolic pathways
Altered metabolism may influence antibiotic uptake, activation, or detoxification
Researchers should employ minimum inhibitory concentration (MIC) assays comparing wild-type and mnmA-deficient strains across a panel of antibiotics, combined with transcriptomic and proteomic analyses to elucidate the specific mechanisms involved.
The L. monocytogenes mnmA enzyme shares significant structural and functional similarities with homologs in other bacterial pathogens, but with important species-specific differences:
Phylogenetic analysis using Maximum Likelihood methods (MEGA X) would reveal evolutionary relationships between these homologs . The conservation of this enzyme across diverse pathogens underscores its fundamental importance in bacterial physiology and potentially represents a broad-spectrum antimicrobial target.
The high conservation of mnmA across bacterial species reflects its fundamental importance in translation fidelity. Evolutionary analysis suggests:
Ancestral Origin: The mnmA gene likely originated in the last universal common ancestor (LUCA), as evidenced by the presence of homologous genes across all three domains of life .
Selective Pressures:
Strong negative selection maintaining core catalytic domains
Positive selection in regions that confer substrate specificity or regulatory functions
Horizontal gene transfer events appear rare, suggesting essential core functionality
Co-evolution with tRNA Substrates:
Coordinated evolution between mnmA and the tRNA genes it modifies
Conservation of recognition elements in tRNA substrates across species
Potential species-specific adaptations in substrate recognition
Functional Divergence:
This evolutionary conservation highlights the critical role of tRNA modifications in translation accuracy and suggests that mnmA represents an ancient and fundamental component of the cellular translation machinery.
The structural features of mnmA offer several promising avenues for antimicrobial drug development:
Targetable Structural Features:
ATP-binding pocket: Design of competitive inhibitors that prevent ATP hydrolysis
tRNA recognition domain: Development of compounds that disrupt enzyme-substrate interactions
Catalytic residues: Identification of covalent inhibitors that irreversibly modify active site residues
Sulfur transfer pathway: Compounds that interfere with the sulfur incorporation mechanism
Drug Development Strategies:
Structure-based virtual screening using homology models
Fragment-based drug discovery targeting specific functional domains
Rational design of transition-state mimetics for the thiolation reaction
Allosteric inhibitors that lock the enzyme in an inactive conformation
Advantages as a Drug Target:
Essential function for bacterial viability and virulence
Absent or structurally distinct from human homologs
Potential for broad-spectrum activity against multiple bacterial pathogens
Inhibition would compromise bacterial adaptation to stress conditions
Experimental Validation Approaches:
Thermal shift assays to confirm compound binding
In vitro enzymatic assays to measure inhibition potency
Cellular assays with reporter systems to monitor translation fidelity
Animal models to assess efficacy against L. monocytogenes infection
Comprehensive analysis of mnmA's impact on the L. monocytogenes transcriptome and proteome requires integrating multiple advanced techniques:
Transcriptomic Approaches:
RNA-Seq: Compare wild-type and mnmA-deficient strains to identify differentially expressed genes
tRNA-Seq: Specialized sequencing to detect changes in tRNA modification patterns
Ribosome profiling: Map ribosome occupancy to identify translation efficiency changes
PARE-seq: Detect aberrant translation events including frameshifting and premature termination
Proteomic Methods:
Quantitative proteomics (iTRAQ or TMT labeling) to identify proteins with altered abundance
Pulse-SILAC to measure protein synthesis rates
Protein turnover analysis to differentiate synthesis versus degradation effects
Post-translational modification profiling to detect compensatory mechanisms
Integrative Analysis:
Multi-omics data integration to correlate transcriptional and translational changes
Codon usage analysis to identify transcripts most affected by mnmA deficiency
Pathway enrichment analysis to identify biological processes impacted
Network analysis to map the regulatory impact of translational disruption
Experimental Validation:
Reporter assays for codon-specific translation efficiency
Western blotting of key identified proteins
Functional assays for specific pathways identified in -omics analyses
This comprehensive approach would reveal both direct effects (on translation of specific codons) and indirect effects (compensatory responses to translation stress) of mnmA function in L. monocytogenes.
The manipulation of mnmA offers promising opportunities for vaccine development:
Rationale for Vaccine Development:
Controlled attenuation: mnmA modification could create strains with reduced virulence but maintained immunogenicity
Translational control: Partial inhibition of mnmA function could limit expression of specific virulence factors
Survival limitation: Engineered mnmA variants could restrict bacterial persistence in specific tissues
Strategies for Vaccine Strain Engineering:
Conditional expression systems: Place mnmA under control of tissue-specific or inducible promoters
Point mutations: Introduce substitutions that partially compromise enzymatic activity
Domain modifications: Engineer chimeric mnmA with altered substrate specificity
Advantages as Vaccine Platforms:
L. monocytogenes naturally elicits robust CD8+ T cell responses
Attenuated strains could safely deliver heterologous antigens
Controlled attenuation would balance safety and immunogenicity
Research Roadmap:
Generate and characterize a panel of mnmA variants with different activity levels
Assess attenuation, tissue tropism, and immune stimulation profiles in animal models
Evaluate protection against lethal challenge with virulent L. monocytogenes
Test delivery of heterologous antigens from other pathogens
CRISPR-based technologies offer powerful approaches to study mnmA function during infection:
In vivo Functional Analysis:
CRISPR interference (CRISPRi): Deploy dCas9-based systems to achieve tunable repression of mnmA expression
Conditional knockout systems: Develop Cre-lox or similar systems for tissue-specific or temporal inactivation
Base editing: Introduce specific point mutations in mnmA to assess structure-function relationships
CRISPR activation (CRISPRa): Upregulate mnmA to evaluate overexpression phenotypes
Spatiotemporal Monitoring:
CRISPR imaging: Tag mnmA with fluorescent reporters to track expression dynamics during infection
Dual reporter systems: Monitor both mnmA expression and bacterial localization simultaneously
Tissue-specific promoters: Drive Cas9 expression only in specific host cell types to achieve selective editing
Host-Pathogen Interaction Studies:
Dual editing: Simultaneously modify bacterial mnmA and host factors to study interactions
Transcriptional recording: Use CRISPR-based recording systems to capture mnmA expression history
Barcoding: Track competitive fitness of different mnmA variants during infection
Technical Considerations:
Delivery systems optimized for in vivo applications
Control measures to prevent off-target effects
Validation strategies to confirm editing efficiency in recovered bacteria
These approaches would provide unprecedented insights into the temporal and spatial requirements for mnmA function during the complex process of L. monocytogenes infection and dissemination .