Recombinant Listeria monocytogenes serotype 4b tRNA-specific 2-thiouridylase mnmA (mnmA)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Listeria monocytogenes Serotype 4b tRNA-specific 2-thiouridylase mnmA (mnmA)

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 .

Characteristics of Listeria monocytogenes Serotype 4b

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 .

Role of mnmA in Bacterial Pathogenesis

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.

Molecular Features of Serotype 4b

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 .

Table 1: Characteristics of Recombinant Listeria monocytogenes Serotype 4b mnmA Protein

CharacteristicDescription
SourceE. coli, Yeast, Baculovirus, Mammalian Cells
TargetListeria monocytogenes Serotype 4b
TypeRecombinant Protein
ApplicationsResearch, Potential Diagnostic/Vaccine Development
PurityVariable depending on production method
StorageTypically at -20°C or -80°C

Table 2: Molecular Features of Listeria monocytogenes Serotype 4b

FeatureDescription
Serotype-Specific GenesgtcA, gltA, gltB
LineagesI and III
Virulence FactorsBeta hemolysin, ability to invade host cells
PathogenicityHigh replication in monocytes/macrophages

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference for your consideration.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
mnmA; trmU; LMOf2365_1531; tRNA-specific 2-thiouridylase MnmA; EC 2.8.1.13
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-371
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
mnmA
Target Protein Sequence
MSTNNSDIRV VVGMSGGVDS SVTAHILKEQ GYDVIGIFMK NWDDTDEFGV CTATEDYDDV IRVANQIGIP YYAVNFEKEY WDKVFTYFLD EYKLGRTPNP DVMCNKEIKF KAFLEHAESL GADYVATGHY AQVKKVGDEI ELLRGVDNNK DQTYFLNQLS QDQLKKVMFP LGGMEKTEVR EIAKKAGLAT ANKKDSTGIC FIGERNFKQF LSEYLPAQPG EMRTLNGEVL GKHDGLMYYT IGQRHGLGIG GDGEPWFVVG KDLKENVLFV EQGFHHETLY SDSLIATDIS FTTNAEKPKT IECTAKFRYR QTDTKVTVHL REDGTAEVVF ADPVRAITPG QAVVFYDGDI CLGGGTIDTV WKNGAKLDYV G
Uniprot No.

Target Background

Function
Catalyzes the 2-thiolation of uridine at the wobble position (U34) of tRNA, resulting in the formation of s2U34.
Database Links
Protein Families
MnmA/TRMU family
Subcellular Location
Cytoplasm.

Q&A

What is the function of tRNA-specific 2-thiouridylase mnmA in Listeria monocytogenes?

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.

How does mnmA contribute to Listeria monocytogenes pathogenicity?

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.

What are the structural characteristics of L. monocytogenes mnmA?

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 .

What are the optimal conditions for expressing recombinant L. monocytogenes mnmA protein?

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

How can researchers effectively design CRISPR/Cas9-based knockout systems for mnmA in Listeria monocytogenes?

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:

    • Construct a plasmid containing the sgRNA, Cas9, and selectable marker

    • Include homology arms (40 bp minimum) flanking the targeted region for homology-directed repair

    • Insert a reporter gene (e.g., EGFP) with a suitable selectable marker (e.g., DHFR) for positive selection

  • 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:

    • Generate a complementation construct containing the wild-type mnmA under its native promoter

    • Include a different selectable marker (e.g., HXGPRT) from the knockout construct

    • Verify successful complementation through PCR, immunofluorescence assay, and Western blot

What methods can be used to evaluate the activity of recombinant mnmA in vitro?

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

How does mnmA contribute to L. monocytogenes dissemination in host organisms?

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:

    • Invasion of intestinal epithelial cells via InlA

    • Intracellular replication essential for cell-to-cell spread

    • Survival within dendritic cells that can transport bacteria to MLN

  • Stress Response Regulation: Proper tRNA modification is crucial for adaptation to stress conditions encountered during:

    • Transition from the intestinal environment to lymphatic vessels

    • Evasion of neutrophil-mediated killing in lymphatic circulation

    • Adaptation to nutrient-limited conditions in the MLN

  • Contribution to Multiple Dissemination Pathways: L. monocytogenes can disseminate via:

    • Cell-associated transport in monocytes or dendritic cells

    • Free-floating bacteria in lymphatic vessels

    • Direct invasion of lymphatic endothelium

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.

What is the impact of environmental stress factors on mnmA expression and activity in L. monocytogenes?

Environmental stress factors likely modulate mnmA expression and activity in L. monocytogenes through multiple regulatory mechanisms:

Environmental StressPredicted Impact on mnmAPhysiological ConsequenceAssessment Method
Acid stress (pH 5.0)UpregulationEnhanced translation fidelity under stressqRT-PCR, Western blot
Oxidative stress (H₂O₂)Initial downregulation followed by compensatory upregulationTemporary translation pause followed by stress adaptationTranscriptomics, proteomics
Nutrient limitationMaintained expressionPrioritized translation of survival genesRibosome profiling
Temperature shift (4°C)Increased activity with stable expressionEnhanced cold adaptation through codon optimizationIn vitro activity assays at various temperatures
Bile exposurePotential inhibition of enzymatic activityReduced virulence in intestinal environmentBile 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.

How does the mnmA-mediated tRNA modification affect antibiotic susceptibility in L. monocytogenes?

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.

How does L. monocytogenes mnmA compare with homologs in other bacterial pathogens?

The L. monocytogenes mnmA enzyme shares significant structural and functional similarities with homologs in other bacterial pathogens, but with important species-specific differences:

OrganismSequence SimilaritySubcellular LocalizationSubstrate SpecificityUnique Features
E. coli~65-70% identityCytoplasmictRNALys, tRNAGlu, tRNAGlnWell-characterized PP-loop domain
Salmonella enterica~60-65% identityCytoplasmictRNALys, tRNAGlu, tRNAGlnAssociated with virulence regulation
Mycobacterium tuberculosis~45-50% identityCytoplasmicLimited substrate rangeContains additional regulatory domains
Staphylococcus aureus~55-60% identityCytoplasmictRNALys, tRNAGlu, tRNAGlnLinked to biofilm formation
Toxoplasma gondii (TgMnmA)~30-35% identityApicoplasttRNALys, tRNAGlu, tRNAGlnEssential for apicoplast function

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.

What is the evolutionary significance of mnmA conservation across bacterial species?

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:

    • While the core function remains conserved (2-thiolation), substrate specificity and regulatory mechanisms may vary

    • In eukaryotes, functional specialization led to compartment-specific variants (e.g., mitochondrial MTU1)

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.

How can structural knowledge of mnmA be leveraged for antimicrobial drug development?

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

What techniques can be used to study the impact of mnmA on the L. monocytogenes transcriptome and proteome?

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.

What are the potential applications of mnmA in developing attenuated L. monocytogenes vaccine strains?

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

How might CRISPR-based technologies be employed to study mnmA function in L. monocytogenes during host infection?

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 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.