Recombinant Nocardia farcinica 50S ribosomal protein L6 (rplF)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Nocardia farcinica 50S Ribosomal Protein L6 (rplF)

Recombinant Nocardia farcinica 50S ribosomal protein L6 (rplF) is a protein derived from the bacterium Nocardia farcinica, a species known for its clinical significance due to its resistance to various antimicrobial agents . The 50S ribosomal protein L6 is an essential component of the large subunit of bacterial ribosomes, playing a crucial role in ribosome assembly and function . This recombinant protein is produced through genetic engineering techniques, allowing for its use in various biochemical and biomedical applications.

Structure and Function of Ribosomal Protein L6

Ribosomal protein L6 is a two-domain protein located on the L7/L12 side of the 50S subunit, forming an L-like structure that bridges between the front and back of the subunit . The N-terminus of L6 interacts with helix 97 of the 23S rRNA, while the C-terminus interacts with the sarcin/ricin loop, which is crucial for the interaction with GTPase translation factors . Mutations in L6 have been shown to affect ribosome assembly, particularly in suppressing defects associated with reduced function of the ribosome assembly factor RbgA .

Production and Handling of Recombinant Nocardia farcinica 50S Ribosomal Protein L6 (rplF)

The recombinant protein is typically produced in a host organism such as Escherichia coli through recombinant DNA technology. After purification, the protein should be reconstituted in deionized sterile water to a concentration of 0.1-1.0 mg/mL. It is recommended to add 5-50% glycerol (final concentration) to enhance stability .

Research Findings and Applications

While specific research on the recombinant Nocardia farcinica 50S ribosomal protein L6 (rplF) is limited, studies on similar proteins highlight their importance in understanding bacterial ribosome assembly and function. The protein's role in ribosome assembly and its interaction with other ribosomal components make it a valuable tool for studying bacterial translation mechanisms .

Table: Key Features of Recombinant Nocardia farcinica 50S Ribosomal Protein L6 (rplF)

FeatureDescription
SourceNocardia farcinica
FunctionEssential for 50S ribosomal subunit assembly and function
StructureTwo-domain protein interacting with 23S rRNA
ProductionRecombinant DNA technology in host organisms like E. coli
HandlingReconstitute in sterile water with glycerol for stability

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on 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 manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rplF; NFA_7890; 50S ribosomal protein L6
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-179
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nocardia farcinica (strain IFM 10152)
Target Names
rplF
Target Protein Sequence
MSRIGKKPIA VPSGVEITID GQHVSVKGPK GTLTHTVAEP ITVTRGEDGQ LEVTRPNDER RNRSLHGLTR TLIANMIEGV TKGYEKKMEI FGVGYRVQAK GSNLEFALGY SHPVPVEAPE GITFAVESPT KFSVSGIDKQ KVGQISAVIH GLRKPDPYKG KGIRYAGEVV RRKVGKTGK
Uniprot No.

Target Background

Function
This protein interacts with 23S rRNA, playing a crucial role in its secondary structure. It is located near the subunit interface at the base of the L7/L12 stalk, in proximity to the peptidyltransferase center's tRNA binding site.
Database Links
Protein Families
Universal ribosomal protein uL6 family

Q&A

What is the genomic context of the rplF gene in Nocardia farcinica?

The rplF gene in Nocardia farcinica is located within its circular chromosome, which contains over 6 million base pairs as revealed through complete genome sequencing . Like most bacterial ribosomal protein genes, rplF is typically found in a conserved operon structure alongside other ribosomal protein genes. This genomic organization is significant for understanding the evolutionary conservation of ribosomal assembly mechanisms. When designing primers for rplF amplification, researchers should consider the flanking regions and potential secondary structures that might affect PCR efficiency.

What expression systems are most effective for producing recombinant N. farcinica rplF protein?

For recombinant expression of N. farcinica rplF, E. coli-based systems (particularly BL21(DE3) strains) offer the highest yield and simplicity for initial studies. Based on experiences with similar ribosomal proteins, expression vectors containing T7 promoters (such as pET series) typically provide robust expression levels. Temperature optimization is critical - expression at lower temperatures (16-25°C) often improves solubility compared to standard 37°C induction. For challenging expressions, consider fusion tags (His6, GST, or MBP) to enhance solubility and facilitate purification. The choice between these systems should be guided by the intended application and required protein folding characteristics.

What purification strategies yield the highest purity of recombinant N. farcinica rplF?

A multi-step purification approach typically yields the highest purity for recombinant N. farcinica rplF. Begin with affinity chromatography (if using a tagged construct), followed by ion exchange chromatography exploiting the protein's theoretical pI. Final polishing via size exclusion chromatography effectively removes aggregates. The table below outlines a recommended purification workflow:

Purification StepMethodBuffer ConditionsExpected YieldPurity Level
AffinityNi-NTA (His-tag)50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10-250 mM imidazole gradient70-80%75-85%
Ion ExchangeQ or SP Sepharose50 mM Tris-HCl pH 8.0, 50-500 mM NaCl gradient60-70%85-95%
Size ExclusionSuperdex 75/20020 mM Tris-HCl pH 7.5, 150 mM NaCl90-95%>95%

Optimizing buffer conditions based on stability testing is essential, as ribosomal proteins can aggregate during concentration steps.

How do mutations in the rplF gene affect N. farcinica antibiotic resistance profiles?

Mutations in the rplF gene can significantly alter ribosomal architecture and consequently impact antibiotic binding and effectiveness. N. farcinica already demonstrates a distinct drug resistance pattern, including resistance to most beta-lactam antibiotics like cefamandole (100%), cefotaxime (100%), and ceftriaxone (80%), as well as resistance to aminoglycosides such as tobramycin (>90%), kanamycin (100%), and gentamicin (100%) . Modifications to the rplF gene may further modulate these patterns by altering ribosome assembly or function. When investigating such mutations, researchers should employ both phenotypic susceptibility testing and molecular techniques to correlate specific mutations with resistance phenotypes.

What structural features distinguish N. farcinica rplF from homologs in other actinomycetes?

The structural distinctions between N. farcinica rplF and its homologs in related actinomycetes center on subtle differences in surface-exposed regions while maintaining a highly conserved core. Key differentiating features include:

  • N-terminal domain variations affecting interactions with ribosomal RNA

  • Surface-exposed loop regions that may influence species-specific ribosomal assembly

  • Distinct electrostatic surface potential distributions related to N. farcinica's unique membrane fatty acid composition, which shows a predominance of straight-chain saturated or monounsaturated fatty acids

Comparative structural analysis through X-ray crystallography or cryo-EM, combined with molecular dynamics simulations, provides the most comprehensive approach to characterizing these differences. When designing experiments, researchers should focus on regions outside the RNA-binding pocket, as these show greater variability while maintaining functional conservation.

How can recombinant N. farcinica rplF be utilized in developing novel diagnostic tools for nocardial infections?

Recombinant N. farcinica rplF offers significant potential for developing diagnostics due to its species-specific epitopes while maintaining sufficient conservation to serve as a broad Nocardia marker. A multi-platform diagnostic approach is recommended:

  • ELISA-based detection: Develop antibodies against unique rplF epitopes for serological detection. Validation studies should include cross-reactivity testing against related Nocardia species, particularly N. nova complex, N. abscessus, N. transvalensis complex, and N. asteroides type VI (N. cyriacigeorgica) .

  • PCR-based identification: Design primers targeting variable regions of the rplF gene. This approach can differentiate N. farcinica from other Nocardia species, addressing the taxonomic complexity highlighted in clinical studies where molecular techniques like PCR restriction enzyme analysis and 16S rRNA sequencing have been necessary for accurate identification .

  • Protein microarray applications: Immobilize recombinant rplF alongside other Nocardia antigens to create comprehensive diagnostic arrays.

When developing these tools, researchers should incorporate controls from clinical isolates with established identification, as misidentification has historically complicated Nocardia taxonomy and diagnostics.

What role does rplF play in N. farcinica virulence and pathogenicity mechanisms?

While primarily a ribosomal structural component, rplF may contribute to N. farcinica virulence through several mechanisms:

  • Moonlighting functions: When exposed on the cell surface or released extracellularly, rplF may interact with host components. Similar moonlighting functions have been demonstrated in other bacteria where ribosomal proteins serve secondary roles in adherence or immune modulation.

  • Translational regulation: Changes in rplF expression or structure may alter the translation efficiency of virulence factors. N. farcinica contains multiple candidate genes for virulence and intrinsic multidrug resistance as identified through genomic analysis .

  • Stress response: Under antibiotic pressure or host immune challenge, altered ribosomal composition (including rplF modifications) may enhance survival.

Research approaches should combine transcriptomic analysis of infection models with protein-protein interaction studies to identify potential host targets. The relationship between rplF and N. farcinica's significant clinical impact, particularly in immunocompromised patients with conditions like cystic fibrosis or leprosy, warrants investigation .

How does post-translational modification of rplF affect ribosome assembly and function in N. farcinica?

Post-translational modifications (PTMs) of rplF can substantially impact ribosome assembly kinetics and translational fidelity in N. farcinica. Common modifications observed in bacterial ribosomal proteins include:

Modification TypePotential Sites on rplFFunctional ConsequenceDetection Method
MethylationLysine residuesAltered rRNA binding affinityMass spectrometry
AcetylationN-terminal and lysine residuesModified ribosome assemblyWestern blot with PTM-specific antibodies
PhosphorylationSerine/threonine residuesRegulated association with ribosomePhosphoproteomic analysis

To study these modifications, researchers should employ a combination of top-down and bottom-up proteomics approaches. Specifically, mass spectrometry analysis of intact protein and peptide fragments from native N. farcinica ribosomes compared to recombinant rplF can reveal differences in modification patterns. Functional studies using reconstituted ribosomes with modified or unmodified rplF can then determine the impact on translation efficiency and fidelity.

What computational approaches best predict interaction networks involving rplF in N. farcinica?

Predicting interaction networks involving rplF in N. farcinica requires integrated computational approaches that combine structural, genomic, and functional data. The most effective computational strategy includes:

  • Homology-based structural modeling: Generate N. farcinica rplF structures using solved ribosomal structures as templates, focusing on interaction interfaces.

  • Molecular docking simulations: Predict interactions with ribosomal RNA, neighboring proteins, and potential antibiotic binding sites.

  • Coevolution analysis: Identify correlated mutations between rplF and potential interaction partners across Actinomycetes.

  • Interactome predictions: Apply machine learning algorithms trained on known bacterial protein-protein interactions to predict novel rplF interaction partners.

When implementing these approaches, researchers should validate computational predictions through experimental methods such as bacterial two-hybrid screening, co-immunoprecipitation, or crosslinking mass spectrometry. The genomic context of N. farcinica, with its large 6-million-base-pair chromosome and two plasmids , provides important constraints for interactome analysis.

What are the optimal conditions for site-directed mutagenesis of the N. farcinica rplF gene?

Site-directed mutagenesis of the N. farcinica rplF gene requires careful optimization to achieve high efficiency. The recommended protocol includes:

  • Primer design considerations: Primers should contain 15-20 nucleotides of perfect match on each side of the desired mutation, with the mutation centered in the primer. GC content should be maintained at 40-60% with a melting temperature of 78-82°C.

  • PCR conditions optimization:

    • Initial denaturation: 98°C for 30 seconds

    • 18 cycles of: 98°C for 10 seconds, 55-65°C for 30 seconds, 72°C for 30 seconds/kb

    • Final extension: 72°C for 10 minutes

  • Template considerations: Use methylated plasmid DNA from a dam+ E. coli strain, allowing selective digestion of parental DNA with DpnI.

  • Verification approach: Sequence the entire rplF coding region after mutagenesis to confirm the presence of desired mutations and absence of secondary mutations.

Critical regions to target for functional studies include the RNA-binding domain and interfaces with adjacent ribosomal proteins based on structural homology modeling.

What analytical techniques provide the most accurate assessment of rplF-mediated ribosome assembly?

Ribosome assembly mediated by rplF can be most accurately assessed through a combination of biophysical and biochemical approaches:

  • Sucrose gradient ultracentrifugation: Provides quantitative analysis of ribosomal subunit ratios and assembly intermediates.

  • Fluorescence-based kinetic assays: Measure real-time assembly using fluorescently-labeled rplF and other components.

  • Cryo-electron microscopy: Visualizes assembly states and conformational changes at near-atomic resolution.

  • Quantitative mass spectrometry: Determines stoichiometry and assembly order of ribosomal components.

The experimental design should include both in vitro reconstitution experiments with purified components and in vivo studies using reporter systems. When analyzing data, researchers should consider the impact of N. farcinica's unique growth characteristics, including its ability to grow at 45°C , which may affect ribosome assembly kinetics compared to model organisms.

How can isothermal titration calorimetry be optimized for studying interactions between N. farcinica rplF and potential binding partners?

Optimizing isothermal titration calorimetry (ITC) for studying rplF interactions requires addressing several technical challenges specific to ribosomal proteins:

  • Buffer composition: Use 20 mM HEPES pH 7.5, 100 mM KCl, 10 mM MgCl₂, and 5 mM β-mercaptoethanol to maintain stability while minimizing buffer mismatch heat effects.

  • Protein concentration determination: Apply amino acid analysis rather than spectrophotometric methods to accurately determine rplF concentration, as ribosomal proteins often have atypical amino acid compositions affecting extinction coefficients.

  • Temperature selection: Conduct experiments at 25°C for initial screening, but include studies at 37°C and 45°C to reflect N. farcinica's growth temperature range .

  • Data analysis considerations: Apply models accounting for potential multiple binding sites when studying rplF-rRNA interactions.

Validation of ITC results should be performed using orthogonal techniques such as surface plasmon resonance or microscale thermophoresis to confirm binding constants and stoichiometry.

How can cryo-electron microscopy advance our understanding of N. farcinica ribosome structure and function?

Cryo-electron microscopy (cryo-EM) offers unprecedented opportunities to elucidate N. farcinica ribosome structure and function at near-atomic resolution. This approach enables:

  • Comparative structural analysis: Direct comparison of N. farcinica ribosomes with other bacterial species to identify unique structural features that may contribute to its distinctive antibiotic resistance profile .

  • Visualization of antibiotic binding: Determination of binding modes for various antibiotics, providing insights into N. farcinica's intrinsic resistance mechanisms.

  • Conformational dynamics: Capturing different functional states of the ribosome to understand species-specific translation regulation.

To maximize success with this approach, researchers should optimize ribosome purification protocols specifically for N. farcinica, considering its unique cell wall composition. Grid preparation techniques may require modification to account for the tendency of nocardial components to aggregate. Data processing should employ classification approaches to identify and analyze structural heterogeneity.

What CRISPR-Cas9 strategies are most effective for genomic modification of rplF in N. farcinica?

Implementing CRISPR-Cas9 genome editing in N. farcinica presents challenges due to its complex cell wall and relatively low transformation efficiency. The most effective strategies include:

  • Delivery system optimization:

    • Electroporation using weakened cell walls (glycine treatment)

    • Conjugation-based transfer from E. coli

    • Mycobacteriophage-based delivery systems adapted for Nocardia

  • CRISPR component selection:

    • SpCas9 with codon optimization for high GC content organisms

    • Single-guide RNA design avoiding the GC-rich regions that predominate in N. farcinica

  • Homology-directed repair templates:

    • 1-2 kb homology arms

    • Selection markers flanked by FRT sites for subsequent removal

  • Screening approach:

    • Colony PCR followed by RFLP analysis

    • Deep sequencing of targeted regions

When designing experiments, researchers should consider the potential essentiality of rplF and prepare conditional knockdown strategies as alternatives to direct gene editing.

How does heterologous expression of N. farcinica rplF in model organisms affect ribosome function and antibiotic sensitivity?

Heterologous expression of N. farcinica rplF in model organisms can provide insights into both ribosomal biology and antibiotic resistance mechanisms. Key experimental considerations include:

  • Expression system selection: E. coli, B. subtilis, and M. smegmatis offer progressively more relevant backgrounds for heterologous expression, with mycobacterial systems providing the closest phylogenetic match.

  • Chimeric ribosome construction: Develop systems where specific ribosomal components can be exchanged between species to isolate the contribution of rplF.

  • Phenotypic analysis:

    • Growth rate determination under various conditions

    • Translational fidelity assays

    • Antibiotic susceptibility testing across multiple classes

The expected outcomes vary by expression system, but common observations include growth defects at high expression levels, altered translation rates, and potentially modified antibiotic sensitivity profiles reflecting N. farcinica's characteristic resistance pattern to multiple beta-lactams and aminoglycosides .

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 2024 Thebiotek. All Rights Reserved.