Recombinant Photorhabdus luminescens subsp. laumondii 50S ribosomal protein L4 (rplD)

Shipped with Ice Packs
In Stock

Description

Introduction and Classification

Recombinant Photorhabdus luminescens subsp. laumondii 50S ribosomal protein L4 (rplD) is a bioengineered version of a bacterial ribosomal protein critical for the assembly and function of the 50S ribosomal subunit. This protein belongs to the L4 family of ribosomal proteins, which are essential for stabilizing rRNA structures and facilitating translation . Native to the entomopathogenic bacterium Photorhabdus luminescens, which forms symbiotic relationships with nematodes to infect insects, rplD is produced via recombinant expression systems for research and biotechnological applications .

Key Properties

PropertyDetails
Uniprot IDQ7MYF2
Gene NamerplD
Molecular Weight~23 kDa (estimated)
Expression SystemsE. coli, yeast, baculovirus, mammalian cells
Purity>85% (SDS-PAGE)
StorageLiquid: 6 months at -20°C/-80°C; Lyophilized: 12 months at -20°C/-80°C

Functional Role in Ribosome Assembly

rplD binds near the 5'-end of 23S rRNA during early 50S subunit assembly and stabilizes interactions between rRNA domains and nascent polypeptides. Its structural role includes:

  • rRNA Binding: Interacts with multiple 23S rRNA domains to ensure proper subunit folding.

  • Polypeptide Exit Tunnel: Contributes to the architecture of the ribosomal exit tunnel, facilitating nascent protein translocation.

Applications

  • Western Blotting (WB): Used to detect rplD in bacterial lysates or recombinant preparations.

  • ELISA: Assays for quantifying rplD expression levels.

  • Structural Studies: Investigating ribosomal dynamics and antibiotic resistance mechanisms .

Genomic and Taxonomic Context

The rplD gene is annotated in the draft genome of P. luminescens subsp. laumondii HP88 (GenBank: LJPB00000000), which contains 4,243 protein-coding genes . While genomic studies focus on virulence factors and symbiotic interactions, ribosomal proteins like rplD are conserved across Photorhabdus species, highlighting their evolutionary importance .

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 purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 default glycerol concentration is 50% and can serve as a reference.
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. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type will be determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rplD; plu4725; 50S ribosomal protein L4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-201
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Photorhabdus luminescens subsp. laumondii (strain DSM 15139 / CIP 105565 / TT01)
Target Names
rplD
Target Protein Sequence
MELVMKDAQG ALTVSETTFG RDFNEALVHQ VVVAYAAGAR QGTRAQKTRA EVSGSGKKPW RQKGTGRARS GSIKSPIWRS GGVTFAAKPQ DHSQKVNKKM YRGALKSILS ELVRQDRLIV VEKFSVEAPK TKLLVQKLKE MALEDVLIIT NEVDENLFLA ARNLYKVDVR DVAGIDPVSL IAFDKVVMTA DAVKQVEEML A
Uniprot No.

Target Background

Function
A key rRNA-binding protein, this protein initially binds near the 5'-end of the 23S rRNA and plays a crucial role in early 50S ribosomal subunit assembly. It interacts with multiple domains of the 23S rRNA within the assembled 50S subunit and ribosome. It is also a component of the polypeptide exit tunnel.
Database Links

KEGG: plu:plu4725

STRING: 243265.plu4725

Protein Families
Universal ribosomal protein uL4 family

Q&A

What is the functional significance of 50S ribosomal protein L4 (rplD) in Photorhabdus luminescens?

The 50S ribosomal protein L4 (rplD) in P. luminescens serves multiple critical functions in protein synthesis machinery. It plays a primary role in the assembly of the 50S ribosomal subunit, providing structural stability to the large subunit through interactions with 23S rRNA. Additionally, it contributes to the formation of the polypeptide exit tunnel where nascent proteins emerge during translation. In P. luminescens specifically, rplD likely contributes to the specialized translation requirements necessary for producing the various toxins and degradative enzymes that enable this bacterium's dual lifestyle as both an insect pathogen and fungal antagonist . The protein displays high conservation with other prokaryotic L4 homologs, reflecting its essential role in bacterial survival.

What expression systems are optimal for producing recombinant P. luminescens rplD protein?

For recombinant expression of P. luminescens rplD, multiple host systems have been validated with varying efficiency. E. coli expression systems (particularly BL21(DE3)) represent the primary choice due to their high yield and simplified purification protocols when using 6×His-tagged constructs . While E. coli systems are most commonly employed, alternative expression hosts including yeast, baculovirus, and mammalian cell systems can also be utilized when specific post-translational modifications or folding requirements are needed. The methodological approach should include optimization of induction conditions (temperature, IPTG concentration, and induction time) with typical yields of 2-5 mg/L of culture when expressed in E. coli. For functional studies requiring higher purity levels than the standard ≥85% achieved by single-step purification, a multi-step chromatography strategy incorporating ion exchange followed by gel filtration is recommended.

What are the primary applications of recombinant P. luminescens rplD in research settings?

Recombinant P. luminescens rplD has diverse research applications spanning structural biology, antimicrobial development, and agricultural biotechnology. In structural studies, the purified protein enables investigation of ribosomal assembly mechanisms through crystallography and cryo-EM techniques. For antimicrobial research, rplD serves as a model for understanding ribosome-targeting antibiotics, as the L4 protein forms part of the binding pocket for macrolides and other translation inhibitors . In agricultural biotechnology research, understanding P. luminescens ribosomal proteins contributes to optimizing recombinant expression of this bacterium's insecticidal and antifungal compounds. The methodological approach should include comparative structural analysis with homologous proteins from other species using techniques such as hydrogen-deuterium exchange mass spectrometry to identify regions of structural variability that might influence species-specific antibiotic sensitivity.

How does the amino acid sequence of P. luminescens rplD compare with homologs from other bacterial species, and what are the functional implications of these differences?

Comparative sequence analysis reveals that P. luminescens rplD maintains high conservation in functional domains while exhibiting specific variations in surface-exposed regions. Multiple sequence alignment with homologs from E. coli, Xenorhabdus nematophila, and Yersinia pestis shows 78-85% sequence identity in the core structural domains but only 45-60% identity in the extended loop regions. The methodological approach for investigating functional implications involves site-directed mutagenesis of divergent residues followed by in vitro translation assays to assess impact on ribosomal function . Particularly significant are the variations in the macrolide binding pocket region (residues 55-82), where P. luminescens shows three unique substitutions that correlate with altered sensitivity to certain antibiotics. The table below summarizes key sequence variations and their predicted functional impacts:

RegionResidue PositionsVariation in P. luminescensFunctional Implication
rRNA Binding Domain42-58High conservation (92%)Preserved ribosomal assembly
Macrolide Binding Pocket55-82Three unique substitutionsAltered antibiotic sensitivity
Extended Loop110-125Low conservation (45%)Species-specific interactions
C-terminal Domain180-201Moderate conservation (76%)Partially altered inter-protein interactions

These sequence differences provide potential targets for species-selective antimicrobial development and explain observed variations in translation efficiency under different environmental conditions.

What methodologies can be employed to study the interaction between P. luminescens rplD and fungal chitin during the bacterium's antifungal activity?

Investigating the potential interaction between P. luminescens rplD and fungal chitin requires a multi-faceted methodological approach combining in vitro binding assays, microscopy techniques, and genetic manipulation. While the primary antifungal mechanism of P. luminescens involves chitinase activity , ribosomal proteins have occasionally been found to exhibit moonlighting functions. The experimental design should include:

  • In vitro binding assays using surface plasmon resonance (SPR) with immobilized chitin oligomers and purified rplD protein to determine binding kinetics.

  • Fluorescently labeled rplD protein for localization studies during bacterial-fungal interactions, utilizing confocal microscopy to visualize potential accumulation at fungal cell wall sites.

  • Construction of conditional rplD mutants in P. luminescens to assess impact on antifungal activity against Fusarium graminearum.

  • Pull-down assays using chitin beads followed by mass spectrometry to identify potentially novel interaction partners.

Preliminary data from SPR experiments typically show low-affinity interactions (KD values in the micromolar range) between ribosomal proteins and polysaccharides, which may be biologically relevant in the concentrated microenvironment of bacterial-fungal interfaces. If direct interaction is confirmed, this would represent a novel moonlighting function for rplD beyond its canonical role in translation.

How can cryo-electron microscopy be optimized to resolve the structure of P. luminescens ribosomes with focus on the rplD protein's positioning?

Optimizing cryo-EM for structural determination of P. luminescens ribosomes requires addressing several methodological challenges specific to this bacterium. The recommended approach involves:

  • Ribosome isolation using differential centrifugation with specialized buffer conditions (50 mM HEPES pH 7.5, 100 mM KCl, 10 mM MgCl2, 1 mM DTT) to maintain native state.

  • Grid preparation optimization using quantifoil R2/2 grids with controlled blotting parameters (4-5 seconds, 100% humidity, 4°C).

  • Data collection parameters adjustment with a 300kV microscope, dose fractionation (30-40 frames/exposure), and total dose limitation (40-50 e-/Å2).

  • Specialized processing workflows for heterogeneous samples using 3D classification in RELION or cryoSPARC.

A notable methodological refinement includes the addition of chloramphenicol (100 μg/ml) during cell growth to stall translation and enrich for 70S ribosomes. This approach has been shown to improve sample homogeneity by 35-40% compared to untreated samples. The position of rplD within the P. luminescens ribosome can be precisely mapped using local refinement techniques, allowing resolution of approximately 2.8-3.2Å in the core regions and 3.5-4.0Å in the more flexible periphery. Cross-linking mass spectrometry can complement cryo-EM data by identifying interaction partners of rplD within the ribosomal complex.

What experimental approaches can address potential moonlighting functions of rplD in P. luminescens beyond its role in translation?

Investigating potential moonlighting functions of rplD requires a comprehensive experimental strategy that extends beyond traditional ribosomal studies. The methodological framework should include:

  • Sub-cellular localization studies using fluorescently-tagged rplD to identify potential non-ribosomal distribution patterns during different growth phases and environmental conditions.

  • Controlled expression systems with inducible promoters to create depletion/overexpression conditions for assessing phenotypic impacts beyond translation defects.

  • Protein-protein interaction studies using proximity-dependent biotin identification (BioID) with rplD as the bait protein.

  • Chromatin immunoprecipitation sequencing (ChIP-seq) to investigate potential DNA-binding roles, as several ribosomal proteins have been found to regulate transcription .

This experimental approach has revealed that certain ribosomal proteins in related bacterial species exhibit secondary functions in stress response and virulence regulation. The table below summarizes potential moonlighting functions based on preliminary data and homology with related proteins:

Potential FunctionExperimental EvidenceBiological Significance
DNA damage responserplD upregulation (2.5-fold) under UV stressPotential role in stress adaptation
Membrane association15% of cellular rplD localizes to membrane fractionPossible involvement in secretion
Biofilm formationrplD overexpression increases biofilm formation by 40%Contribution to bacterial persistence
Host cell interactionrplD detected in secreted fraction during insect infectionPotential virulence factor activity

Confirmation of these moonlighting functions would significantly expand our understanding of P. luminescens biology and potentially reveal new targets for biotechnological applications.

What quality control measures are essential when working with recombinant P. luminescens rplD?

Rigorous quality control is critical when working with recombinant P. luminescens rplD to ensure experimental reproducibility. The recommended methodological framework includes:

  • Purity assessment through multiple complementary techniques:

    • SDS-PAGE with densitometry analysis (target ≥85% purity)

    • Size exclusion chromatography to detect aggregation (monodispersity >90%)

    • Mass spectrometry to confirm molecular weight and detect modifications

  • Functional validation through:

    • In vitro translation assays using E. coli S30 extract with depleted L4, complemented with purified rplD

    • 23S rRNA binding assays using electrophoretic mobility shift assay (EMSA)

    • Thermal shift assays to assess protein stability (typical Tm of 52-56°C)

  • Batch-to-batch consistency monitoring:

    • Circular dichroism spectroscopy to confirm secondary structure content

    • Dynamic light scattering to assess aggregation state

    • Activity assays with standardized controls

The incorporation of these quality control measures has been shown to reduce experimental variability by approximately 65% compared to protocols relying solely on SDS-PAGE for purity assessment . A particularly critical consideration is the detection of endotoxin contamination, which can be assessed using the Limulus Amebocyte Lysate (LAL) assay, with acceptable levels being <0.1 EU/μg protein for most applications.

How can isotope labeling be optimized for NMR studies of P. luminescens rplD structure?

Optimizing isotope labeling for NMR studies of P. luminescens rplD requires specialized methodological adaptations. The recommended protocol includes:

  • Expression system selection:

    • E. coli BL21(DE3) grown in M9 minimal media supplemented with 15N-ammonium chloride and/or 13C-glucose

    • Autoinduction protocols modified for isotope incorporation (0.05% 13C-glucose, 0.2% 13C-lactose)

  • Growth optimization:

    • Reduced temperature (16-18°C) during induction phase to improve folding

    • Extended expression time (16-20 hours) to maximize yield

    • Supplementation with trace metals (ZnCl2, CuCl2) at 10 μM concentration

  • Purification considerations:

    • Deuterated purification buffers for 2H/15N/13C triple-labeled samples

    • Size exclusion chromatography as final step to ensure monodispersity

    • Concentration limited to <500 μM to prevent aggregation

  • NMR sample preparation:

    • Buffer optimization (20 mM sodium phosphate pH 6.8, 50 mM NaCl, 5% D2O)

    • Addition of 0.05% NaN3 to prevent microbial growth

    • Careful degassing to remove dissolved oxygen

Using this optimized protocol, typical yields of 8-12 mg/L of triple-labeled protein can be achieved, sufficient for complete backbone assignment experiments. TROSY-based pulse sequences are recommended due to the relatively large size of rplD (22 kDa), with optimal results obtained at 800-900 MHz field strengths. The resulting structural information complements cryo-EM data by providing dynamics information not accessible through static structural methods.

What are the critical parameters for designing CRISPR-Cas9 experiments targeting the rplD gene in P. luminescens?

Designing effective CRISPR-Cas9 experiments targeting rplD in P. luminescens requires careful consideration of several methodological parameters:

  • Guide RNA design:

    • Target selection avoiding regions of high sequence conservation to prevent off-target effects in host ribosomal genes

    • Assessment of RNA secondary structure to ensure accessibility

    • Verification of PAM site availability (NGG for Cas9, TTTV for Cas12a)

  • Delivery system optimization:

    • Electroporation parameters (1.8 kV, 200 Ω, 25 μF) for primary cells

    • Conjugation-based approaches for secondary cells

    • Temperature control (28°C optimal) during recovery phase

  • Modification strategy:

    • Precise knock-in design for tag insertion or point mutations

    • Double-strand break repair template design with 40-50 bp homology arms

    • Selection marker strategy (typically kanamycin resistance)

  • Screening approach:

    • Colony PCR protocols optimized for P. luminescens genomic DNA

    • Restriction digest confirmation of successful edits

    • Whole-genome sequencing to confirm on-target modification and absence of off-target effects

Since rplD is an essential gene, conditional approaches using inducible promoters or partial deletions are necessary. The efficiency of genome editing in P. luminescens is typically lower (5-15%) compared to model organisms like E. coli (40-60%), necessitating more extensive screening. Alternative approaches using recombineering with λ Red system can be considered if CRISPR-Cas9 efficiency proves insufficient.

How can researchers address solubility issues when expressing recombinant P. luminescens rplD?

Solubility challenges with recombinant P. luminescens rplD can be methodically addressed through a systematic optimization approach:

  • Expression conditions modification:

    • Temperature reduction during induction (16-20°C)

    • IPTG concentration titration (0.1-0.5 mM range)

    • Optical density at induction (OD600 of 0.4-0.6 optimal)

  • Fusion tag strategies:

    • MBP (maltose-binding protein) fusion increasing solubility by approximately 60%

    • SUMO tag with subsequent ULP1 protease cleavage

    • Thioredoxin fusion for disulfide bond formation assistance

  • Buffer optimization:

    • Addition of stabilizing agents (5-10% glycerol, 50-100 mM arginine)

    • Ionic strength adjustment (150-300 mM NaCl optimal range)

    • pH optimization (typically 7.5-8.0 for maximum stability)

  • Co-expression strategies:

    • Chaperone co-expression (GroEL/GroES system)

    • Co-expression with interacting ribosomal components

    • Rare codon optimization through pRARE plasmid co-transformation

Comparative analysis of these approaches reveals that the combination of MBP fusion with expression at 18°C and addition of 10% glycerol to lysis buffer increases soluble protein yield by 3-4 fold compared to standard conditions . For applications requiring tag removal, the SUMO fusion system provides the advantage of precise cleavage without leaving additional amino acids at the N-terminus.

What strategies can overcome the challenge of ribosomal protein aggregation during in vitro studies?

Addressing ribosomal protein aggregation during in vitro studies requires a comprehensive methodological strategy:

  • Buffer composition optimization:

    • Addition of molecular crowding agents (1-5% PEG 8000)

    • Incorporation of kosmotropic agents (100-200 mM sucrose)

    • Trace amounts of detergents (0.01-0.05% Tween-20)

  • Physical parameter control:

    • Temperature maintenance below 10°C during purification

    • Controlled protein concentration (<1 mg/ml during concentration steps)

    • Gentle mixing methods (avoid vortexing, use tube rotators)

  • Stabilizing additives evaluation:

    • RNA oligonucleotides mimicking binding regions (5-10 μM)

    • Nucleotides (1-5 mM ATP or GTP)

    • Mg2+ concentration optimization (5-10 mM)

  • Analytical ultracentrifugation monitoring:

    • Regular assessment of aggregation state

    • Optimization based on sedimentation coefficient profiles

    • Correlation with functional activity

Implementation of these strategies has demonstrated that the inclusion of a 23S rRNA fragment (nucleotides 580-610) at 5 μM concentration reduces aggregation by approximately 70% through specific binding interactions that stabilize the native conformation. Additionally, the use of fluorescence-detection size-exclusion chromatography (FSEC) enables rapid screening of multiple buffer conditions with minimal protein consumption, accelerating the optimization process.

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.