Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The specific tag type is determined during production. To request a specific tag, please indicate this during ordering, and we will prioritize its development.
This protein facilitates GTP-dependent binding of aminoacyl-tRNA to the ribosomal A-site during protein biosynthesis.
KEGG: plu:plu0432
STRING: 243265.plu0432
Elongation Factor Tu 1 (tuf1) in Photorhabdus luminescens functions primarily during protein synthesis by mediating the entry of aminoacyl tRNA into the ribosome's free site. This critical role in translational elongation is essential for the bacterium's protein synthesis machinery. As in other bacteria, EF-Tu is one of the most abundant proteins and plays a central role in maintaining translational accuracy and efficiency . In the context of P. luminescens, which has a complex lifecycle involving symbiosis with nematodes and pathogenicity to insects, efficient protein synthesis is crucial for adapting to these diverse environments and producing the various adhesins, toxins, proteases, and other virulence factors encoded in its genome .
While specific sequence comparisons are not provided in the search results, EF-Tu is generally highly conserved among bacterial species due to its fundamental role in protein synthesis. In P. luminescens, the tuf1 gene would be expected to show high sequence homology with other enterobacteria such as Escherichia coli. The bacterial EF-Tu proteins share homology with their eukaryotic counterparts (eEF-1A), though with distinct structural differences . This conservation reflects the evolutionary importance of the translational machinery. For experimental design purposes, researchers should consider these homologies when designing primers for cloning or when selecting antibodies for detection, as cross-reactivity might occur between different bacterial EF-Tu proteins.
For recombinant expression of P. luminescens tuf1, E. coli-based expression systems are generally most suitable due to their genetic similarity as enterobacteria. Based on established protocols for recombinant protein expression, the following systems would be recommended:
| Expression System | Advantages | Considerations |
|---|---|---|
| E. coli BL21(DE3) | High expression levels, well-characterized | May require optimization of induction parameters |
| E. coli Rosetta | Enhanced expression of proteins with rare codons | Useful if P. luminescens tuf1 contains rare codons |
| E. coli Arctic Express | Low-temperature expression capability | Beneficial if protein aggregation occurs at higher temperatures |
| E. coli SHuffle | Enhanced disulfide bond formation | Useful if proper folding requires disulfide bonds |
The choice of vector system should include strong promoters like T7 and appropriate affinity tags (His-tag, GST) to facilitate purification. Temperature optimization is particularly important given P. luminescens' temperature sensitivity . Expression trials at different temperatures (16°C, 25°C, 30°C) should be conducted to determine optimal conditions for soluble protein production.
A multi-step purification approach is recommended for obtaining high-purity, active recombinant tuf1:
Affinity chromatography: If expressing His-tagged tuf1, immobilized metal affinity chromatography (IMAC) using Ni-NTA resin provides effective initial purification.
Ion exchange chromatography: As a second step, anion exchange chromatography can remove remaining contaminants based on charge differences.
Size exclusion chromatography: A final polishing step to separate monomeric tuf1 from aggregates and other size-based impurities.
Buffer composition is critical for maintaining activity. Based on general EF-Tu purification protocols, the following buffer system is recommended:
| Purification Stage | Buffer Composition |
|---|---|
| Cell Lysis | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 5 mM MgCl₂, 1 mM DTT, 10% glycerol, protease inhibitors |
| IMAC | 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 5 mM MgCl₂, 20-250 mM imidazole (gradient) |
| Ion Exchange | 20 mM Tris-HCl pH 7.5, 50-500 mM NaCl (gradient), 5 mM MgCl₂, 1 mM DTT |
| Size Exclusion | 20 mM HEPES pH 7.5, 100 mM KCl, 10 mM MgCl₂, 1 mM DTT |
The presence of Mg²⁺ is particularly important as EF-Tu is a GTPase that requires this cofactor for proper folding and activity.
The GTPase activity of recombinant tuf1 can be measured using several complementary approaches:
Malachite green assay: This colorimetric method quantifies inorganic phosphate released during GTP hydrolysis. A standard reaction mixture would contain:
1-5 μM purified recombinant tuf1
0.5-1 mM GTP
50 mM Tris-HCl pH 7.5
10 mM MgCl₂
100 mM KCl
Coupled enzymatic assay: This approach links GTP hydrolysis to NADH oxidation, which can be monitored spectrophotometrically at 340 nm.
Radioactive GTP hydrolysis assay: Using [γ-³²P]GTP to directly measure released radiolabeled phosphate.
For comparative analysis, researchers should include control reactions without protein, with heat-inactivated tuf1, and potentially with EF-Tu from model organisms like E. coli. Activity measurements at different temperatures would be particularly relevant given P. luminescens' temperature-sensitive physiology .
To investigate tuf1's interaction with aminoacyl-tRNAs, researchers can employ several techniques:
Filter binding assays: Using radiolabeled aminoacyl-tRNAs to quantify binding to purified tuf1 under various conditions.
Surface plasmon resonance (SPR): Provides real-time binding kinetics between immobilized tuf1 and flowing aminoacyl-tRNAs.
Microscale thermophoresis (MST): Measures interactions in solution with minimal sample consumption.
Electrophoretic mobility shift assay (EMSA): Visualizes tuf1-tRNA complex formation.
Experimental design should consider the effect of temperature, nucleotides (GTP vs. GDP), and Mg²⁺ concentration. Given P. luminescens' adaptation to both insect and nematode environments , comparing aminoacyl-tRNA binding at temperatures relevant to these contexts (15-28°C for soil/nematode and 28-37°C for insect hosts) would provide valuable insights into potential temperature-dependent functional adaptations.
Beyond its canonical role in translation, bacterial EF-Tu proteins can have additional functions including:
Cell surface association and potential role in host interactions
Potential involvement in stress responses
Possible roles in protein folding
To investigate these functions in P. luminescens tuf1, researchers could:
Conduct pull-down assays with recombinant tuf1 followed by mass spectrometry to identify interaction partners in P. luminescens cell lysates.
Generate conditional tuf1 expression mutants to assess phenotypic changes beyond growth defects, particularly examining effects on:
Perform immunolocalization studies to determine if tuf1 localizes to the cell membrane or is secreted under certain conditions, which might suggest roles in host-pathogen interactions.
Given P. luminescens' complex lifestyle involving symbiosis with nematodes and pathogenicity to insects , tuf1 might have adapted specialized functions related to these ecological niches.
P. luminescens transitions between nematode symbiosis and insect pathogenicity, environments with different temperatures and physiological conditions . To investigate tuf1's potential role in these adaptations:
Compare tuf1 expression levels and post-translational modifications between P. luminescens grown under conditions mimicking nematode gut (approximately 25°C) versus insect hemolymph (28-37°C) using qRT-PCR, Western blotting, and proteomics.
Analyze the GTPase activity and aminoacyl-tRNA binding properties of recombinant tuf1 under varying temperature and pH conditions relevant to different host environments.
Investigate potential interactions between tuf1 and host factors using pull-down assays with nematode or insect tissue extracts followed by mass spectrometry.
Generate point mutations in tuf1 based on sequence comparisons with other bacterial EF-Tu proteins and assess their effects on P. luminescens growth and virulence under different environmental conditions.
This research could reveal whether tuf1 serves as an environmental sensor that helps facilitate P. luminescens' adaptation to different hosts, particularly in relation to the temperature-restricted lifestyle documented in these bacteria .
P. luminescens exists in two phenotypically different cell types: primary (1°) cells that are symbiotic with nematodes and secondary (2°) cells that remain in soil after insect infection . To investigate potential differences in tuf1 between these phenotypes:
Compare tuf1 expression levels and potential isoforms between 1° and 2° cells using RNA-seq and proteomic approaches.
Examine potential post-translational modifications of tuf1 in both cell types, which might alter its function or localization.
Assess tuf1 activity (GTPase and aminoacyl-tRNA binding) in protein extracts from both cell types to identify functional differences.
Investigate whether tuf1 interacts with different protein partners in 1° versus 2° cells through co-immunoprecipitation followed by mass spectrometry.
Determine if tuf1 overexpression or depletion affects the switch between 1° and 2° phenotypes.
This research direction could reveal whether translational control through tuf1 contributes to the phenotypic plasticity of P. luminescens, which is particularly relevant given that 2° cells specifically interact with plant roots and protect plants from phytopathogenic fungi .
Solubility challenges are common when expressing recombinant proteins. For P. luminescens tuf1, consider these approaches:
| Challenge | Solution Strategies | Implementation Details |
|---|---|---|
| Inclusion body formation | Lower expression temperature | Induce at 16-20°C for 16-24 hours |
| Reduce inducer concentration | Use 0.1-0.5 mM IPTG instead of 1 mM | |
| Use solubility-enhancing tags | Try MBP, SUMO, or TrxA fusion tags | |
| Protein aggregation | Optimize buffer conditions | Include stabilizing agents (5-10% glycerol, 1 mM DTT) |
| Co-express with chaperones | Use plasmids encoding GroEL/GroES or DnaK/DnaJ/GrpE | |
| Low expression levels | Codon optimization | Adapt rare codons to E. coli preference |
| Try different promoter systems | Compare T7, tac, and araBAD promoters |
Based on P. luminescens' temperature sensitivity , expression at lower temperatures might be particularly suitable for maintaining proper folding of tuf1. Additionally, ensuring the presence of Mg²⁺ in all buffers is crucial for proper folding of GTPases like EF-Tu.
When performing functional assays with recombinant tuf1, several technical challenges may arise:
Low GTPase activity:
Ensure protein is properly folded (circular dichroism spectroscopy)
Verify nucleotide binding using fluorescent nucleotide analogs
Check for inhibitory contaminants in protein preparation
Optimize Mg²⁺ concentration (typically 5-10 mM)
Ensure GTP quality and freshness
Poor aminoacyl-tRNA binding:
Verify tRNA aminoacylation status
Ensure correct buffer conditions (particularly K⁺ and Mg²⁺ concentrations)
Check GTP presence and concentration
Optimize temperature conditions based on P. luminescens physiology
Data interpretation challenges:
Include appropriate positive controls (e.g., E. coli EF-Tu)
Perform parallel assays under different temperature conditions (15-37°C)
Consider the influence of post-translational modifications that might occur in vivo but not in recombinant systems
Given P. luminescens' adaptation to different environments (nematode gut, insect hemolymph, soil) , conducting assays at temperatures relevant to these various habitats would provide more ecologically relevant functional insights.
When faced with contradictory results in tuf1 research, consider these methodological approaches:
Validate protein identity and integrity:
Perform mass spectrometry to confirm protein sequence
Check for proteolytic degradation using SDS-PAGE and Western blotting
Verify proper folding using circular dichroism or thermal shift assays
Control for experimental variables:
Employ complementary techniques:
If in vitro and in vivo results conflict, use conditional expression systems in P. luminescens
Combine biochemical assays with structural approaches (X-ray crystallography, cryo-EM)
Use site-directed mutagenesis to create tuf1 variants that can test specific hypotheses
Consider biological context:
Resolving contradictions often requires integrating multiple lines of evidence and considering the unique biological context of P. luminescens as both a symbiont and pathogen.