Recombinant Escherichia coli O45:K1 Methionyl-tRNA formyltransferase (fmt)

Shipped with Ice Packs
In Stock

Description

Overview of Methionyl-tRNA Formyltransferase (FMT)

Methionyl-tRNA formyltransferase (FMT) is a critical enzyme in prokaryotic protein synthesis, responsible for formylating the initiator methionyl-tRNA (tRNA<sup>fMet</sup>). This modification ensures the initiation of translation with formylmethionine, a hallmark of bacterial and organellar protein synthesis . In Escherichia coli, FMT is encoded by the fmt gene and is essential for normal cell growth .

2.1. Enzyme Architecture

  • Domains: FMT comprises an N-terminal Rossmann fold domain for binding N<sup>10</sup>-formyltetrahydrofolate (THF) and a C-terminal β-barrel domain for tRNA binding .

  • Catalytic Mechanism: The enzyme transfers a formyl group from THF to the methionine moiety of tRNA<sup>fMet</sup>, enabling ribosomal recognition during initiation .

2.2. Key Mutational Studies

  • C-Terminal Domain: Deletion of 18–80 amino acids from the C-terminus severely impairs activity, highlighting its role in tRNA binding .

  • Suppressor Mutations: Substitutions like G41R in the linker region restore formylation activity in defective tRNA substrates .

MutationImpact on ActivityReference
C-terminal deletionsLoss of tRNA binding
G41R substitutionEnhanced tRNA formylation
A89L/A172L (human)Reduced catalytic efficiency

3.1. Heterologous Systems

  • Yeast Expression: Recombinant E. coli FMT has been expressed in Saccharomyces cerevisiae, leading to formylation of cytoplasmic tRNA<sup>fMet</sup> and growth defects, which are rescued by co-expression of E. coli deformylase (DEF) .

  • Purification: His-tagged FMT variants (wild-type and mutants like R42L) are purified via affinity chromatography for biochemical assays .

4.1. Antibiotic Targeting

FMT is absent in humans, making it a potential target for novel antibiotics. Inhibitors disrupting formylation could selectively impair bacterial protein synthesis .

4.2. Role in Virulence

  • Pathogen Adaptation: Horizontal gene transfer of O-antigen clusters in E. coli O45:K1 enhances serum resistance and meningeal invasion . Although FMT itself is not directly linked to virulence genes, its essential role in translation underscores its indirect contribution to pathogen fitness .

Research Gaps and Future Directions

  • Strain-Specific Studies: No published data explicitly characterize FMT in E. coli O45:K1. Comparative analyses with archetypal strains (e.g., K12) are needed to elucidate potential structural or functional adaptations.

  • Therapeutic Exploration: High-resolution structural data (e.g., PDB: 1FMT ) could guide the design of FMT inhibitors tailored to meningitis-causing strains.

Q&A

What is Methionyl-tRNA formyltransferase (fmt) and what is its role in bacterial protein synthesis?

Methionyl-tRNA formyltransferase (fmt) catalyzes the formylation of methionyl-tRNA (Met-tRNA) to formylmethionyl-tRNA (fMet-tRNA), which serves as the initiator tRNA for protein synthesis in bacteria, mitochondria, and chloroplasts. This formylation step is crucial for efficient translation initiation in bacteria. The formyl group attached to methionine plays a dual role: it acts as a positive determinant for the initiation factor IF2 and as a negative determinant for the elongation factor EF-Tu . This biochemical distinction helps ensure that formylated Met-tRNA participates exclusively in translation initiation rather than elongation.

The fmt enzyme typically uses 10-formyl-tetrahydrofolate (10-CHO-THF) as the formyl donor, though recent research has demonstrated that 10-formyldihydrofolate (10-CHO-DHF) can also serve as an alternative substrate for formyl group donation . Deletion of the fmt gene causes severe growth retardation in Escherichia coli and Streptococcus pneumoniae, highlighting its physiological importance, though interestingly such deletion is less detrimental in Pseudomonas aeruginosa and Staphylococcus aureus .

How does E. coli O45:K1 differ from other E. coli K1 strains in terms of fmt characteristics?

E. coli O45:K1 represents a distinct serotype within the E. coli K1 group that has been identified as predominant in neonates with E. coli infections . Comparative genomic hybridization studies have demonstrated that E. coli K1 strains isolated from cerebrospinal fluid (CSF) can be categorized into two distinct groups based on their profiles of virulence factors, lipoproteins, proteases, and outer membrane proteins .

While the specific characteristics of fmt in E. coli O45:K1 have not been extensively characterized compared to other K1 strains, the genomic differences between K1 groups suggest potential variations in translation-related enzymes like fmt. Group 1 and Group 2 E. coli K1 strains differ in their secretion systems, with Group 2 strains containing open reading frames encoding the type III secretion system apparatus, while Group 1 strains predominantly utilize the general secretory pathway . These differences in secretion mechanisms may potentially influence the expression, regulation, or post-translational modifications of fmt, though direct experimental evidence for this would require targeted comparative studies.

What is the substrate specificity of fmt and how can it be experimentally determined?

The fmt enzyme exhibits notable substrate specificity with regard to both its tRNA acceptor and formyl donor substrates. Current research has established the following substrate profile:

Substrate TypePrimary SubstrateAlternative SubstrateDetection Method
AcceptorMethionyl-tRNA (Met-tRNA)None identifiedRadioisotope labeling, HPLC
Formyl Donor10-formyl-tetrahydrofolate (10-CHO-THF)10-formyldihydrofolate (10-CHO-DHF)LC-MS/MS analysis

To experimentally determine substrate specificity, several methodological approaches are recommended:

  • Radioisotope-based assays: Using [14C]-labeled formyl donors to quantify the incorporation of formyl groups into Met-tRNA. This method provides high sensitivity but requires special handling of radioactive materials.

  • HPLC separation: Distinguishing formylated from non-formylated Met-tRNA by reverse-phase HPLC and quantifying by UV absorbance (typically at 254 nm).

  • Mass spectrometry verification: The formation of byproducts like dihydrofolate (DHF) can be verified by LC-MS/MS analysis, as demonstrated in recent studies where 10-CHO-DHF was confirmed as an alternative substrate .

  • Comparative enzyme kinetics: Determining Km and kcat values for different potential substrates to assess relative efficiency of utilization.

Recent in vitro and in vivo approaches have demonstrated that fmt can utilize 10-CHO-DHF as an alternative formyl group donor, expanding our understanding of the enzyme's substrate flexibility .

What expression systems are optimal for producing recombinant fmt from E. coli O45:K1?

The optimal expression of recombinant fmt from E. coli O45:K1 requires careful consideration of expression systems and conditions. Based on protocols developed for similar bacterial enzymes, the following methodological approach is recommended:

Expression System Design:

  • Vector selection: pET series vectors containing T7 promoter systems offer high-level, inducible expression appropriate for fmt

  • Affinity tags: N-terminal His6-tag facilitates purification while generally preserving enzymatic activity

  • Host strain: E. coli BL21(DE3) or Rosetta(DE3) strains are preferred, with the latter providing additional tRNAs for rare codons that may be present in O45:K1 sequences

Optimization Parameters:

  • Induction conditions: IPTG concentration (0.1-0.5 mM), temperature (16-25°C), and duration (4-16 hours) require optimization

  • Growth media: Rich media (LB, 2xYT) for high yield; minimal media for isotope labeling if structural studies are planned

  • Temperature shift: Growing cells at 37°C until OD600 reaches 0.6-0.8, then reducing to 18°C prior to induction enhances proper folding

Purification Strategy:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin

  • Secondary purification: Size exclusion chromatography to ensure homogeneity

  • Activity preservation: Including 5-10% glycerol and reducing agents (DTT, 1-5 mM) in purification buffers to maintain enzyme activity

Experimental validation of expression conditions should include activity assays to ensure that the recombinant enzyme retains full catalytic function, as improper folding can result in inactive protein despite high expression levels.

How can site-directed mutagenesis be utilized to investigate the catalytic mechanism of fmt?

Site-directed mutagenesis represents a powerful approach for investigating the catalytic mechanism and structure-function relationships of fmt. A strategic approach includes:

Target Selection for Mutagenesis:

  • Conserved residues: Identified through multiple sequence alignments of fmt across bacterial species

  • Predicted active site residues: Based on structural data or homology modeling

  • Substrate binding determinants: Residues interacting with tRNA or formyl donor

  • Structural elements: Residues maintaining essential secondary structure elements

Experimental Design:

  • Alanine scanning: Systematically replacing targeted residues with alanine to assess their importance

  • Conservative substitutions: Replacing residues with chemically similar amino acids to probe specific properties

  • Charge inversions: Changing charged residues to test electrostatic interactions

Functional Analysis of Mutants:

Mutation TypeExpected Effect if Residue is CriticalExperimental Readout
Active site residuesDramatic decrease in kcatEnzyme kinetics assay
Substrate binding residuesIncreased Km valuesBinding affinity studies
Structural residuesAltered protein stabilityThermal denaturation assays

Studies with human mitochondrial MTF (a homolog of bacterial fmt) have demonstrated that mutations in certain conserved residues significantly affect enzyme activity. Particularly, the strategic positioning of small aliphatic amino acids has been shown to be required for normal MTF function . Similar approaches could reveal critical residues in E. coli O45:K1 fmt.

For comprehensive mechanistic insights, combining mutagenesis with structural studies and computational modeling provides the most robust approach to understanding fmt catalysis.

What are the optimal enzymatic assay conditions for measuring fmt activity from recombinant E. coli O45:K1?

Establishing optimal enzymatic assay conditions is critical for accurate measurement of fmt activity. The following methodological considerations should be implemented:

Buffer Composition:

  • pH range: 7.5-8.0 (typically using Tris-HCl or HEPES buffer)

  • Salt concentration: 50-100 mM KCl or NaCl to maintain ionic strength

  • Divalent cations: 5-10 mM Mg2+ is essential for tRNA stability

  • Reducing agents: 1-5 mM DTT or β-mercaptoethanol to maintain thiol groups

Substrate Preparation:

  • Met-tRNA substrate: Generated by aminoacylation of purified tRNA^Met using methionyl-tRNA synthetase

  • Formyl donor: Freshly prepared 10-CHO-THF (or alternatively 10-CHO-DHF) to avoid degradation

Reaction Conditions:

  • Temperature: 30-37°C (with temperature optimization recommended)

  • Time course: Linear range typically within 2-10 minutes

  • Enzyme concentration: Adjusted to achieve 10-20% substrate conversion

Detection Methods:

  • Radiochemical assay: Using [14C]-labeled formyl donor with quantification by liquid scintillation counting

  • HPLC-based detection: Separating and quantifying formylated vs. non-formylated Met-tRNA

  • Mass spectrometry: For high-sensitivity detection of reaction products

Kinetic Analysis:

  • Substrate concentration range: 0.1-10× Km values for accurate determination of kinetic parameters

  • Michaelis-Menten analysis: To determine Km and Vmax for both tRNA and formyl donor substrates

  • pH profile analysis: To identify optimal pH and potential catalytic residues

When comparing fmt variants or studying inhibitors, consistent reaction conditions are essential for meaningful comparisons. Control reactions lacking either substrate or enzyme should be included to account for background and non-enzymatic reactions.

How does fmt activity contribute to E. coli O45:K1 pathogenicity in neonatal meningitis?

The relationship between fmt activity and E. coli O45:K1 pathogenicity in neonatal meningitis involves several interconnected mechanisms, though direct experimental evidence specifically linking fmt to virulence in this strain remains limited. Multiple lines of evidence suggest potential pathogenic contributions:

Translational Efficiency and Virulence Factor Expression:
E. coli K1 is the leading gram-negative organism causing neonatal meningitis . As fmt is crucial for efficient translation initiation in bacteria, it likely influences the expression levels of virulence factors required for meningeal invasion and survival in cerebrospinal fluid. E. coli K1 strains have been categorized into two groups based on virulence factor profiles , suggesting that translation efficiency differences could contribute to virulence heterogeneity.

Growth Rate and Infection Dynamics:
Deletion of fmt causes severe growth retardation in E. coli , indicating that optimal fmt activity is required for bacterial fitness. In the context of neonatal infection, growth rate can be critical for overwhelming host defenses before adaptive immunity develops.

Stress Response and Adaptation:
During infection, bacteria encounter various stresses including nutrient limitation and host defense mechanisms. Efficient translation initiation through fmt activity may be particularly important under these stress conditions, allowing for rapid adaptation through synthesis of stress-response proteins.

Potential Differential Expression:
E. coli O45:K1 has been identified as predominant in neonates with E. coli infections , suggesting potential unique characteristics. Comparative studies of fmt expression levels or activity between virulent and avirulent strains could reveal whether fmt upregulation contributes to enhanced virulence in this serotype.

Despite these potential mechanisms, a direct causal relationship requires further investigation through approaches such as:

  • Construction of fmt conditional mutants in E. coli O45:K1

  • Virulence studies in appropriate neonatal meningitis models

  • Transcriptomic and proteomic analyses comparing wildtype and fmt-deficient strains

Can fmt serve as a viable target for antimicrobial development against pathogenic E. coli strains?

The potential of fmt as an antimicrobial target against pathogenic E. coli strains requires evaluation across multiple criteria:

Target Validation:

CriterionAssessmentEvidence
EssentialityPartialDeletion causes severe growth retardation in E. coli but not complete lethality
ConservationHighPresent across bacterial species with conserved mechanism
Selective ToxicityModerateAbsent in cytoplasmic translation in eukaryotes but present in mitochondria
DruggabilityFavorableWell-defined catalytic site amenable to small molecule inhibition

Pros of fmt as a Target:

  • Fmt inhibition would significantly impair bacterial growth rates, potentially allowing host defenses to clear infection

  • The absence of fmt in cytoplasmic translation in eukaryotes provides a basis for selective toxicity

  • The unique biochemistry of bacterial translation initiation offers opportunities for specific inhibitor design

  • Partial essentiality may reduce selection pressure for resistance compared to bactericidal targets

Challenges and Considerations:

  • The viability of fmt deletion mutants, albeit with impaired growth, suggests that resistance could emerge

  • Presence of mitochondrial MTF in humans raises potential for off-target effects

  • The variable importance of fmt across bacterial species may limit broad-spectrum applications

  • Developing inhibitors that can penetrate the bacterial cell membrane may present challenges

Strategic Approaches:

  • Development of fmt inhibitors as adjuvants to existing antibiotics rather than standalone therapies

  • Design of selective inhibitors exploiting structural differences between bacterial fmt and human mitochondrial MTF

  • Targeting fmt in combination with folate metabolism, as FolD-deficient and fmt-overexpressing strains show increased sensitivity to trimethoprim

The recent demonstration that fmt can utilize alternative substrates suggests flexibility in the enzyme that would need to be considered in inhibitor design. While not an ideal standalone target due to partial essentiality, fmt represents a promising component of combination therapies or for situations where bacteriostatic activity is sufficient.

How does fmt expression correlate with antibiotic resistance profiles in clinical E. coli O45:K1 isolates?

The relationship between fmt expression and antibiotic resistance profiles in clinical E. coli O45:K1 isolates remains incompletely characterized, but several mechanistic connections warrant investigation:

Folate Pathway Connection:
Research has demonstrated that FolD-deficient mutants and fmt-overexpressing strains show increased sensitivity to trimethoprim (TMP) compared to Δfmt strains . This finding establishes a link between fmt activity and antibiotic susceptibility through the folate metabolism pathway. Trimethoprim inhibits dihydrofolate reductase (DHFR), affecting the availability of tetrahydrofolate derivatives required for fmt activity.

Translation Efficiency and Resistance Gene Expression:
Fmt's role in translation initiation efficiency may affect the expression levels of resistance determinants. Many resistance mechanisms require substantial protein synthesis, including:

  • Efflux pump components

  • β-lactamases like the extended-spectrum β-lactamases (ESBLs) found in some pathogenic E. coli

  • Alternative metabolic enzymes that bypass antibiotic targets

Stress Response and Persistence:
Efficient translation initiation may be particularly important under antibiotic stress conditions, allowing bacteria to rapidly synthesize stress-response proteins. This could potentially influence:

  • Development of tolerance states

  • Formation of persister cells

  • Adaptation to antibiotic pressure

Experimental Approaches to Investigate Correlations:

  • Transcriptomic analysis: Compare fmt expression levels across clinical isolates with different resistance profiles

  • Gene knockdown studies: Assess how reduced fmt expression affects minimum inhibitory concentrations for various antibiotics

  • Reporter constructs: Measure how fmt expression changes in response to antibiotic exposure

  • Clinical correlations: Analyze fmt expression or genetic variants in relation to treatment outcomes

The transfer of extended-spectrum beta-lactamase (ESBL)-producing E. coli between patients has been documented , highlighting the clinical relevance of understanding resistance mechanisms. Whether fmt plays a direct role in such resistance remains to be established through targeted research.

What methodological challenges exist in studying fmt structure-function relationships?

Several significant methodological challenges complicate the study of fmt structure-function relationships:

Substrate Complexity and Availability:
The fmt enzyme requires two complex substrates—Met-tRNA and 10-CHO-THF—neither of which is commercially available in forms suitable for enzymatic studies. Researchers must prepare these substrates through:

  • In vitro transcription of tRNA followed by aminoacylation with methionyl-tRNA synthetase

  • Chemical synthesis or enzymatic generation of 10-CHO-THF, which is unstable and light-sensitive

Structural Characterization Difficulties:
Obtaining high-resolution structural data presents several challenges:

  • Crystallization difficulties due to flexible regions within the protein

  • The need for co-crystallization with substrates or substrate analogs to capture relevant conformations

  • Challenges in maintaining the integrity of bound tRNA during structural studies

  • Limited availability of strain-specific structural information for E. coli O45:K1 fmt

Enzyme Assay Complexity:
Developing reliable activity assays involves several technical challenges:

  • Distinguishing between formylated and non-formylated Met-tRNA

  • Achieving linear reaction kinetics for accurate measurement

  • Accounting for potential product inhibition effects

  • Developing high-throughput compatible assays for inhibitor screening

Genetic Manipulation Constraints:
The growth defects associated with fmt deletion complicate genetic studies:

  • Difficulties in generating stable knockout strains

  • Potential compensatory mutations arising during strain construction

  • Phenotypic analysis complicated by growth rate differences

  • Challenges in complementation studies

Future Methodological Approaches:

Addressing these methodological challenges will require interdisciplinary approaches combining expertise in enzymology, structural biology, molecular genetics, and analytical biochemistry.

What emerging technologies could advance research on fmt as an antimicrobial target?

Several emerging technologies show promise for advancing research on fmt as an antimicrobial target:

1. Structural Biology Innovations:

  • Cryo-electron microscopy (cryo-EM): Enables visualization of fmt-substrate complexes without crystallization

  • Micro-electron diffraction (MicroED): Allows structure determination from nanocrystals

  • Room-temperature crystallography: Captures enzymatically relevant conformational states

  • Time-resolved structural methods: Captures enzyme dynamics during catalysis

2. Advanced Computational Approaches:

  • AI-driven protein structure prediction: Tools like AlphaFold2 can predict fmt structures with high accuracy

  • Molecular dynamics simulations: Reveal binding mechanisms and conformational changes

  • Machine learning for inhibitor prediction: Identifies novel chemical scaffolds with potential inhibitory activity

  • Quantum mechanics/molecular mechanics (QM/MM): Elucidates detailed catalytic mechanisms

3. Chemical Biology Tools:

  • Activity-based protein profiling: Identifies active site residues and monitors inhibitor engagement

  • Fragment-based drug discovery: Efficient screening of chemical space for fmt inhibitors

  • Photo-crosslinking probes: Captures transient enzyme-substrate interactions

  • Click chemistry approaches: Enables in situ labeling of fmt interaction partners

4. Genetic Technologies:

  • CRISPR interference (CRISPRi): Allows tunable repression of fmt expression

  • Directed evolution: Generates fmt variants with altered properties for mechanistic studies

  • Genome-wide interaction screens: Identifies genetic interactions with fmt

  • Single-cell technologies: Reveals heterogeneity in fmt expression and function

5. Translational Research Approaches:

  • Pharmacokinetic/pharmacodynamic modeling: Predicts in vivo efficacy of fmt inhibitors

  • Hollow fiber infection models: Simulates physiological conditions for testing fmt inhibitors

  • Ex vivo human tissue models: Evaluates safety and efficacy without animal testing

  • Biomarker development: Identifies indicators of successful fmt targeting

Integration of these technologies could overcome current research limitations and accelerate the development of fmt as an antimicrobial target, particularly for pathogens like E. coli O45:K1 where traditional antibiotics face resistance challenges.

How might fmt research contribute to understanding broader bacterial translation regulation mechanisms?

Research on fmt has significant potential to illuminate broader bacterial translation regulation mechanisms through several interconnected avenues:

Translation Initiation Control Mechanisms:
The formylation of Met-tRNA by fmt represents a key control point in bacterial translation initiation. Studies of fmt can reveal:

  • How bacteria modulate translation initiation rates under different environmental conditions

  • The interplay between formylation efficiency and ribosome binding site accessibility

  • Potential regulatory mechanisms affecting fmt activity as a means to control global protein synthesis

Metabolic Integration with Protein Synthesis:
Fmt utilizes 10-CHO-THF from the folate metabolism pathway , establishing a direct link between central metabolism and translation. This connection may reveal:

  • How bacteria coordinate protein synthesis with metabolic state

  • The impact of nutritional limitations on translation initiation

  • Metabolic adaptations that maintain translation under stress conditions

Evolutionary Insights:
The variable essentiality of fmt across bacterial species provides a natural experiment in translation evolution:

  • Comparative studies can reveal how some bacteria have adapted to function efficiently without formylation

  • Analysis of compensatory mechanisms in fmt-deficient strains can uncover novel translation regulation pathways

  • Evolutionary trajectories of translation systems can be traced by examining fmt conservation patterns

Stress Response Integration:
Translation initiation is a critical control point during stress responses. Fmt research could reveal:

  • How bacteria modulate formylation efficiency during various stresses

  • The relationship between translation initiation and stress-response protein expression

  • Potential stress-specific regulation of fmt activity or expression

Clinical and Translational Implications:
The role of fmt in pathogenic strains like E. coli O45:K1 connects translation regulation to virulence:

  • Comparative studies of fmt expression or activity between virulent and avirulent strains

  • Analysis of how host environments influence fmt function

  • Identification of pathogen-specific translation regulation mechanisms

By integrating fmt research into a broader systems biology context, we can advance beyond viewing fmt as merely a housekeeping enzyme and understand its role in the complex regulatory networks that govern bacterial physiology, adaptation, and pathogenesis.

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.