Recombinant Nocardia farcinica (Dimethylallyl)adenosine tRNA methylthiotransferase MiaB (miaB), partial, is a recombinant protein derived from the bacterium Nocardia farcinica. This enzyme is involved in the modification of tRNA molecules, specifically in the addition of a methylthiol group to the dimethylallyl moiety of adenosine in tRNA. The MiaB enzyme plays a crucial role in maintaining translational fidelity by enhancing the interaction between tRNA and ribosomes during protein synthesis.
Species Origin: Nocardia farcinica (strain IFM 10152).
Purity: Greater than 85% as determined by SDS-PAGE.
Uniprot No.: Q5YT08.
Sequence: The sequence of the recombinant MiaB protein includes amino acids such as MTRARSYEIR TFGCQMNVHD SERLSGLLED AGYVKAAPGA TADLVVFNTC AVRENADNKL YGTLGHLAPV KAGRPGMQIA VGGCLAQKDR DTVVRKAPWV DVVFGTHNIG SLPVLLERAR HNEQAQVEIL ESLEAFPSTL PAKRESAYAG .
MiaB enzymes in bacteria are generally involved in the modification of tRNA molecules, which is essential for maintaining the accuracy of protein synthesis. In Nocardia farcinica, this enzyme could play a similar role, ensuring that the bacterium can efficiently synthesize proteins necessary for its survival and virulence. Nocardia farcinica is an opportunistic pathogen known for causing nocardiosis, a disease that affects various tissues in humans and animals .
tRNA Modification: MiaB modifies tRNA by adding a methylthiol group, which enhances the interaction between tRNA and ribosomes, thus improving translational fidelity.
Protein Synthesis: Essential for the accurate synthesis of proteins involved in bacterial metabolism and virulence.
Antibacterial Targets: Understanding the role of MiaB in Nocardia farcinica could lead to the development of novel antibacterial strategies targeting tRNA modification pathways.
Biotechnology: The recombinant enzyme could be used in biotechnological applications, such as enhancing protein synthesis in industrial microorganisms.
Function | Description |
---|---|
tRNA Modification | Adds a methylthiol group to tRNA, enhancing translational fidelity. |
Protein Synthesis | Essential for accurate protein synthesis, affecting bacterial metabolism and virulence. |
Note: We will prioritize shipping the format currently in stock. If you have specific format requirements, please specify them during order placement.
Note: All proteins are shipped with standard blue ice packs unless otherwise requested. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Catalyzes the methylthiolation of N6-(dimethylallyl)adenosine (i6A), resulting in the formation of 2-methylthio-N6-(dimethylallyl)adenosine (ms2i6A) at position 37 in tRNAs that recognize codons beginning with uridine.
KEGG: nfa:NFA_38350
STRING: 247156.nfa38350
MiaB is a bifunctional radical-S-adenosylmethionine (radical-AdoMet) enzyme that catalyzes the posttranscriptional methylthiolation of N-6-isopentenyladenosine in tRNAs . In Nocardia farcinica, MiaB plays a critical role in tRNA modification, which affects protein translation fidelity and efficiency. While the specific function in N. farcinica hasn't been fully characterized, research on MiaB from other organisms suggests it contributes to translational accuracy by enabling proper codon-anticodon interactions. N. farcinica contains multiple candidate genes for virulence and intrinsic multidrug resistance within its circular chromosome of over 6 million base pairs , and MiaB-catalyzed tRNA modifications may influence expression of these virulence factors.
MiaB contains two distinct [4Fe-4S]²⁺,¹⁺ clusters that are essential for its enzymatic activity:
Iron-Sulfur Cluster | Coordination | Function |
---|---|---|
Cluster 1 | Coordinated by three conserved cysteines in the radical-AdoMet motif (e.g., Cys150, Cys154, Cys157 in T. maritima MiaB) | Initiates radical chemistry by cleaving AdoMet |
Cluster 2 | Coordinated by three N-terminal conserved cysteines (e.g., Cys10, Cys46, Cys79 in T. maritima MiaB) | Proposed to act as a sacrificial sulfur donor for the thiolation reaction |
Both clusters are essential for full catalytic activity, as reconstituted forms of MiaB containing two [4Fe-4S] clusters demonstrate significantly higher activity than those with a single cluster . The clusters have similar UV-visible absorption, resonance Raman, and Mössbauer properties but differ in their redox properties and EPR characteristics when reduced . This difference is critical for their sequential roles in the catalytic mechanism.
Multiple complementary spectroscopic techniques should be employed for comprehensive characterization:
UV-visible absorption spectroscopy: Provides initial confirmation of [4Fe-4S] cluster incorporation, typically showing a broad peak around 400-420 nm.
Resonance Raman spectroscopy: Enables identification of Fe-S stretching modes characteristic of [4Fe-4S] clusters.
Mössbauer spectroscopy: Critical for distinguishing between different types of iron-sulfur clusters and determining their oxidation states. This technique has been successfully applied to T. maritima MiaB to characterize both [4Fe-4S]²⁺ clusters .
EPR spectroscopy: Essential for analyzing the reduced [4Fe-4S]¹⁺ clusters, which exhibit distinct EPR signatures. The two clusters in MiaB exhibit different EPR properties when reduced, providing a method to distinguish between them .
Circular dichroism: Useful for monitoring structural changes associated with cluster binding and substrate interactions.
For optimal results, these analyses should be performed on highly purified protein samples under anaerobic conditions to prevent oxidative degradation of the iron-sulfur clusters.
Successfully producing active recombinant N. farcinica MiaB requires careful attention to several key factors:
Expression System Design:
Express in E. coli BL21(DE3) or similar strains under the control of a regulated promoter (T7 or araBAD)
Co-express with iron-sulfur cluster assembly proteins (ISC or SUF system components) to enhance cluster incorporation
Culture under microaerobic or anaerobic conditions to minimize cluster degradation
Supplement growth media with iron and cysteine to support cluster formation
Purification Protocol:
All steps should be conducted anaerobically in a glove box or using degassed buffers with reducing agents
Initial capture using affinity chromatography (His-tag or GST-tag)
Secondary purification by ion exchange chromatography
Final polishing by size exclusion chromatography
Maintain 1-5 mM DTT or β-mercaptoethanol throughout purification
Reconstitution of Iron-Sulfur Clusters:
The enzymatic activity of MiaB is directly dependent on proper incorporation of both [4Fe-4S] clusters. For optimal reconstitution:
Incubate purified MiaB under strictly anaerobic conditions with excess Fe²⁺/Fe³⁺ (typically ferrous ammonium sulfate)
Add sulfide source (sodium sulfide)
Include a reducing agent (DTT or sodium dithionite)
Allow reconstitution to proceed for 3-4 hours at 25°C
Remove excess reconstitution components by desalting or dialysis
This approach, similar to that used for T. maritima MiaB , has been shown to yield enzyme with both [4Fe-4S] clusters properly incorporated and significantly higher activity.
Distinguishing N. farcinica proteins from mycobacterial proteins is particularly important given the clinical and bacteriological similarities between Nocardia and Mycobacterium species . Several complementary approaches can be employed:
PCR-Based Differentiation:
PRA-hsp65 method can differentiate Nocardia from Mycobacterium species
Nocardia lacks the BstEII restriction site present in mycobacterial hsp65 genes
HaeIII restriction patterns for Nocardia differ from those described for Mycobacterium species
Mass Spectrometry Analysis:
MALDI-TOF MS profiles can distinguish between Nocardia and Mycobacterium proteins
Targeted LC-MS/MS of tryptic digests can identify species-specific peptides
Immunological Methods:
Western blotting using antibodies specific to N. farcinica MiaB
ELISA assays with species-specific epitope recognition
These approaches are particularly important in clinical and research settings where misidentification can lead to improper treatment strategies or experimental confounds .
A robust experimental design for measuring MiaB activity should include:
Substrate Preparation:
Isolate or synthesize the appropriate tRNA substrate containing N-6-isopentenyladenosine (i⁶A)
Ensure substrate purity through HPLC purification and mass spectrometry verification
Reaction Conditions:
Anaerobic environment to maintain [4Fe-4S] cluster integrity
Buffer system at pH 7.5-8.0 with physiologically relevant ionic strength
Include S-adenosylmethionine as methyl donor
Add reducing system (dithionite or flavodoxin/flavodoxin reductase/NADPH)
Include source of sulfur for thiolation (likely from the second iron-sulfur cluster)
Analysis of Products:
HPLC separation of modified and unmodified tRNAs
Mass spectrometry to confirm addition of both methyl and thiol groups
Radioisotope labeling using ¹⁴C-labeled SAM to quantify methyl transfer
Controls:
No-enzyme control
Heat-inactivated enzyme control
Variant enzyme lacking one or both iron-sulfur cluster binding sites
By systematically varying reaction components and conditions, researchers can determine the kinetic parameters and cofactor requirements specific to N. farcinica MiaB.
When confronting contradictory data regarding MiaB activity and antimicrobial resistance in N. farcinica, researchers should implement a systematic analytical approach:
Common Sources of Contradictions:
Strain heterogeneity - N. farcinica isolates show variable drug resistance profiles
Growth condition variations affecting MiaB expression
Methodological differences in activity assays or susceptibility testing
Analytical Framework:
Verify strain identity: Confirm N. farcinica identification using molecular methods like 16S rRNA sequencing and PRA-hsp65, as Nocardia is frequently misidentified as Mycobacterium .
Standardize susceptibility testing: N. farcinica has a specific drug susceptibility pattern characterized by:
Examine experimental conditions: Parameters including temperature, pH, and media composition can significantly affect both MiaB activity and antimicrobial resistance gene expression.
Statistical analysis: Apply multivariate analysis to identify confounding variables and control for their effects.
Complementary approaches: Combine biochemical activity assays with genetic approaches (gene knockout/complementation) to establish causal relationships.
When reporting contradictory findings, researchers should clearly document all experimental conditions and strain characteristics to facilitate reproducibility and resolution of discrepancies.
Optimizing recombinant N. farcinica MiaB expression and activity requires a structured experimental design approach:
Sequential Experimentation Process:
Screening Design: Begin with fractional factorial designs to identify the most significant factors affecting MiaB expression and activity from potential variables including:
Temperature
Inducer concentration
Media composition
Harvest time
Iron supplementation
Oxygen levels
Modeling Design: Once key factors (typically 2-5) are identified, implement higher-resolution designs to examine interactions and potential curvature in responses .
Response Surface Methodology: For final optimization, use central composite or Box-Behnken designs to model the response surface and identify optimal conditions.
Practical Implementation:
For 2-5 factors, use resolution IV or V designs that can detect main effects and two-factor interactions
For designs with 6-9 factors, consider folding to add runs that increase precision and power
Place replicates and axial points in separate blocks to account for different experimental conditions
Analysis Considerations:
Use backward elimination method for model refinement
Account for block effects in statistical models
Calculate effect sizes in standard deviation units to ensure biological relevance
This structured approach significantly increases the probability of identifying optimal conditions while minimizing the number of experiments required.
The relationship between N. farcinica MiaB activity and pathogenesis involves several interconnected mechanisms:
Translational Fidelity and Stress Response:
MiaB catalyzes the methylthiolation of specific tRNAs, which affects translational accuracy and efficiency. In pathogenic contexts, this modification likely influences the expression of virulence factors and stress response proteins. N. farcinica contains multiple candidate genes for virulence within its 6-million-base-pair circular chromosome , and their regulated expression may depend on properly modified tRNAs.
Diagnostic Challenges and Treatment Implications:
N. farcinica is often misidentified as Mycobacterium species due to similar clinical presentation and bacteriological characteristics . This misidentification can lead to inappropriate treatment, as N. farcinica exhibits specific drug resistance patterns:
Antimicrobial Class | Susceptibility Pattern | Clinical Implication |
---|---|---|
Beta-lactams | Resistant to most, including cefamandole (100%), cefotaxime (100%), ceftriaxone (80%) | First-line beta-lactams likely ineffective |
Aminoglycosides | Resistant to tobramycin (>90%), kanamycin (100%), gentamicin (100%) | Most aminoglycosides ineffective except amikacin |
Other agents | Susceptible to amikacin (100%), sulfamethoxazole (100%), imipenem (82%), ciprofloxacin (88%) | These agents are preferred treatment options |
This intrinsic multidrug resistance pattern may be influenced by translational regulation through tRNA modifications catalyzed by MiaB.
Clinical Case Evidence:
In documented cases, patients with nocardiosis were initially misdiagnosed with tuberculosis due to similar presentation. After failing standard TB therapy, further investigation revealed Nocardia infection . This treatment failure pattern was observed in 31.2% of patients initially treated for TB but actually infected with Nocardia . The relationship between MiaB activity and these clinical outcomes represents an important area for further investigation.
Distinguishing between MiaB enzymatic activity and the presence of mixed infections requires a multifaceted approach:
Molecular Identification Techniques:
PCR-based methods targeting the hsp65 gene with restriction enzyme analysis can differentiate Nocardia from Mycobacterium, as Nocardia lacks the BstEII restriction site .
16S rRNA gene sequencing provides definitive species identification, with most N. farcinica clinical isolates showing 100% homology with the ATCC type strain .
PRA-hsp65 method can specifically identify N. farcinica, differentiating it from other Nocardia species and Mycobacterium .
Microbiological Approaches:
Colony morphology assessment: N. farcinica typically forms rough, pigmented colonies with filamentous aerial hyphae .
Lysozyme resistance testing: N. farcinica is resistant to lysozyme, which helps distinguish it from some other actinomycetes .
Growth at 45°C: N. farcinica can grow at this temperature, unlike many other Nocardia species .
Biochemical Assays:
Utilization of acetamide as nitrogen and carbon source
Acid production from rhamnose
Mixed Infection Analysis:
Three documented cases have shown mixed infections of different Nocardia and Mycobacterium species in immunocompetent patients . In these cases, specific identification of both pathogens is critical, as treatment approaches differ significantly. When TB treatment fails or patient condition worsens despite adequate therapy, pulmonary nocardiosis should be considered in the differential diagnosis .
By implementing these complementary approaches, researchers can accurately distinguish between MiaB activity in N. farcinica and potential mixed infections with Mycobacterium species, leading to more appropriate experimental designs and treatment strategies.
Investigating the role of the second [4Fe-4S] cluster as a potential sulfur donor requires sophisticated experimental designs:
Site-Directed Mutagenesis Studies:
Create variants with mutations in the cysteine residues that coordinate the second [4Fe-4S] cluster (likely Cys10, Cys46, and Cys79, based on T. maritima MiaB) .
Characterize these variants spectroscopically to confirm selective disruption of the second cluster while maintaining the radical-AdoMet cluster.
Assess enzymatic activity to determine if methylation still occurs without thiolation.
Isotope Labeling Experiments:
Reconstitute the enzyme with ³⁵S-labeled iron-sulfur clusters.
Perform activity assays and track the transfer of the labeled sulfur to the tRNA substrate.
Analyze products by mass spectrometry to determine the origin of the incorporated sulfur atom.
Spectroscopic Time-Course Analysis:
Monitor changes in the spectroscopic properties of both [4Fe-4S] clusters during the reaction.
Use rapid freeze-quench techniques coupled with EPR and Mössbauer spectroscopy to capture intermediates.
Compare with other radical-AdoMet enzymes involved in thiolation reactions, such as biotin synthase and lipoate synthase .
Structural Studies:
Determine high-resolution crystal structures of N. farcinica MiaB in various states:
Substrate-free
Bound to tRNA substrate
In the presence of AdoMet
After completion of the reaction
Analyze structural changes in the second [4Fe-4S] cluster during catalysis.
This comprehensive approach would provide definitive evidence regarding whether the second [4Fe-4S] cluster serves as a sacrificial sulfur donor, similar to what has been proposed for other radical-AdoMet enzymes involved in thiolation reactions .
Designing a systematic drug discovery campaign targeting N. farcinica MiaB requires:
Target Validation Studies:
Generate conditional MiaB knockdown strains in N. farcinica.
Assess impact on virulence in cellular and animal infection models.
Determine essentiality under various stress and growth conditions.
High-Throughput Screening Strategy:
Develop a fluorescence-based or coupled enzymatic assay amenable to high-throughput format.
Screen diverse chemical libraries against purified recombinant N. farcinica MiaB.
Implement counterscreens to eliminate compounds that generally disrupt iron-sulfur proteins.
Rational Design Approach:
Focus on compounds that can:
Interfere with AdoMet binding
Disrupt [4Fe-4S] cluster integrity
Prevent substrate recognition
Utilize homology modeling based on known MiaB structures from other species.
Employ molecular docking and virtual screening methodologies.
Lead Optimization Cascade:
Biochemical characterization:
Determine mechanism of inhibition (competitive, noncompetitive, uncompetitive)
Measure binding kinetics using surface plasmon resonance
Assess specificity by testing against related radical-AdoMet enzymes
Cellular studies:
Measure antimicrobial activity against N. farcinica
Determine activity against drug-resistant clinical isolates
Evaluate toxicity against mammalian cells
Pharmacological optimization:
Improve physicochemical properties for bacterial penetration
Enhance stability and reduce metabolism
Optimize selectivity for bacterial versus human enzymes