Recombinant Leptospira interrogans serogroup Icterohaemorrhagiae serovar copenhageni Elongation factor Tu (tuf)

Shipped with Ice Packs
In Stock

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 purchasing 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 advance notification 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. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations 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. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. To request a specific tag, please inform us, and we will prioritize its development.
Synonyms
tuf; LIC_12875; Elongation factor Tu; EF-Tu
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-401
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Leptospira interrogans serogroup Icterohaemorrhagiae serovar copenhageni (strain Fiocruz L1-130)
Target Names
tuf
Target Protein Sequence
MAKEKFDRSK PHLNVGTIGH VDHGKTTLTA AITTTLAKAI GGKNKAVAYD QIDNAPEEKA RGITIATSHQ EYETANRHYA HVDCPGHADY VKNMITGAAQ MDAAILVVSA TDGPMPQTKE HILLARQVGV PYVIVFINKA DMLAADERAE MIEMVEMDVR ELLNKYSFPG DTTPIVHGSA VKALEGDESE IGMPAILKLM EALDTFVPNP KRVIDKPFLM PVEDVFSITG RGTVATGRVE QGVLKVNDEV EIIGIRPTTK TVVTGIEMFR KLLDQAEAGD NIGALLRGTK KEEIERGQVL AKPGSITPHK KFAAEVYVLT KDEGGRHTPF INNYRPQFYF RTTDVTGVCN LPNGVEMVMP GDNVSLTVEL ISPIAMDKGL KFAIREGGRT IGSGVVAEIT E
Uniprot No.

Target Background

Function
This protein facilitates the GTP-dependent binding of aminoacyl-tRNA to the ribosomal A-site during protein biosynthesis.
Database Links
Protein Families
TRAFAC class translation factor GTPase superfamily, Classic translation factor GTPase family, EF-Tu/EF-1A subfamily
Subcellular Location
Cytoplasm.

Q&A

What is the molecular structure and primary function of Leptospira Elongation Factor Tu?

Elongation Factor Tu (EF-Tu) is a highly conserved 43 kDa protein originally known for its role in protein synthesis where it delivers aminoacyl-tRNAs to the ribosome. In Leptospira interrogans serovar copenhageni, EF-Tu has been identified as a "moonlighting protein" with additional functions beyond protein synthesis. The protein is highly conserved among diverse Leptospira species, sharing 99-100% identity at the amino acid level among pathogenic strains. When compared with the non-pathogenic L. biflexa Patoc1, the EF-Tu protein still maintains 86% sequence identity, indicating its evolutionary conservation across the genus .

How is EF-Tu distributed among different Leptospira species and serovars?

EF-Tu is ubiquitously distributed across Leptospira species. Immunoblot analysis using anti-EF-Tu serum has demonstrated the presence of a 43 kDa protein, corresponding to the expected size of native EF-Tu, in all tested pathogenic serovars including Panama, Javanica, Tarassovi, Cynopteri, Copenhageni, Pomona, and Shermani. Importantly, this protein is also present in the non-pathogenic saprophytic L. biflexa serovar Patoc .

The conservation of EF-Tu across pathogenic and non-pathogenic Leptospira suggests it provides essential functions beyond virulence. While its moonlighting activities may contribute to pathogenesis in virulent strains, the presence of EF-Tu in non-pathogenic strains indicates its fundamental importance to leptospiral biology regardless of pathogenic potential.

How does Leptospira EF-Tu compare to EF-Tu in other bacterial species?

EF-Tu from Leptospira interrogans shares significant sequence homology with EF-Tu proteins from other bacterial species, but with distinct variations reflecting evolutionary adaptations. The table below summarizes the functional roles of surface EF-Tu in different microorganisms and their sequence similarity to L. interrogans Copenhageni L1-130:

MicroorganismFunction (associated with surface localization)% identity to L. interrogans Copenhageni L1-130% similarity to L. interrogans Copenhageni L1-130
Pseudomonas aeruginosaInteraction with human complement Factor H and plasminogen72%86%
Francisella novicidaEliciting of inflammatory cytokine response in macrophages70%85%
Listeria monocytogenesInteraction with plasminogen72%84%
Mycoplasma pneumoniaeInteraction with fibronectin68%80%
Lactobacillus johnsoniiBinding to human intestinal cells and mucin66%80%

This comparison indicates that while EF-Tu is highly conserved across bacterial species (>65% identity), the protein has evolved specialized moonlighting functions in different bacteria while maintaining its primary role in protein synthesis .

What is the mechanism by which Leptospira EF-Tu contributes to immune evasion?

Leptospira EF-Tu contributes to immune evasion through two primary mechanisms: interaction with Factor H (FH) and binding of plasminogen.

First, EF-Tu acquires FH, a 150-kDa plasma protein that inhibits the alternative pathway of complement activation. When bound to EF-Tu, FH maintains its cofactor activity for Factor I (FI)-mediated cleavage of C3b, as demonstrated by the presence of C3b cleavage fragments after incubation with FI. This prevents the deposition of C3b on the leptospiral surface, inhibiting opsonization and formation of the membrane attack complex .

Second, EF-Tu binds human plasminogen in a dose-dependent manner, primarily through lysine residues, as evidenced by reduced binding in the presence of ε-aminocaproic acid. Once bound, plasminogen is converted to enzymatically active plasmin, which can degrade the complement protein C3b, further contributing to complement evasion. The dual activity of acquiring both FH and plasminogen makes EF-Tu a significant contributor to leptospiral immune evasion strategies .

It's worth noting that while EF-Tu is present in non-pathogenic leptospires, pathogenic strains have additional complement evasion mechanisms, including other surface proteins like LenA, LenB, LigA, LigB, and LcpA, which are absent in non-pathogenic species.

How does surface-localized EF-Tu facilitate tissue invasion by Leptospira?

Surface-localized EF-Tu facilitates tissue invasion by Leptospira through multiple mechanisms:

  • ECM Binding: EF-Tu mediates interactions with extracellular matrix components including collagen I and IV, cellular and plasma fibronectin, laminin, and elastin. These interactions allow Leptospira to adhere to host tissues, a critical initial step in colonization .

  • Plasminogen Activation: EF-Tu-bound plasminogen is converted to active plasmin, which can degrade fibrinogen and potentially other ECM components. This proteolytic activity may create breaches in tissue barriers, facilitating bacterial dissemination .

  • Complement Evasion: By inhibiting complement activation through FH acquisition and C3b degradation, EF-Tu helps Leptospira avoid immune clearance during tissue invasion and dissemination .

  • Fibrinogen Interaction: Direct binding to fibrinogen, combined with the ability to degrade it through activated plasmin, may interfere with coagulation and contribute to hemorrhagic manifestations seen in severe leptospirosis .

These mechanisms work synergistically to enhance leptospiral invasiveness, allowing the bacteria to penetrate tissues and disseminate throughout the host.

Why did EF-Tu fail as a vaccine candidate despite its surface exposure and conservation?

Despite EF-Tu's surface exposure and high conservation across Leptospira species, it failed to elicit protection in hamsters challenged with lethal doses of L. interrogans serovar Copenhageni L1-130 . Several factors may explain this failure:

  • Structural Constraints: The portions of EF-Tu exposed on the bacterial surface may be limited or conformationally distinct from the recombinant protein used for immunization, resulting in antibodies that cannot effectively target native surface-exposed EF-Tu.

  • Immunological Evasion: The high conservation of EF-Tu across bacterial species suggests that hosts may have evolved tolerance to this protein to prevent autoimmune reactions to commensal bacteria, limiting robust immune responses.

  • Functional Redundancy: Pathogenic Leptospira express multiple virulence factors that can compensate for the neutralization of EF-Tu. For example, other proteins like LenA, LenB, LigA, LigB, and LcpA also contribute to complement evasion .

  • Insufficient Immunogenicity: As a highly conserved bacterial protein, EF-Tu may not be sufficiently immunogenic to elicit protective antibody titers, especially if tolerance mechanisms are involved.

Understanding these limitations is crucial for researchers developing vaccines against Leptospira, suggesting that effective vaccines may require combinations of antigens targeting multiple virulence mechanisms simultaneously.

What are the recommended protocols for purifying recombinant Leptospira EF-Tu for functional studies?

For purification of recombinant Leptospira EF-Tu, the following methodological approach is recommended:

  • Gene Cloning:

    • Amplify the tuf gene from L. interrogans serovar Copenhageni genomic DNA using high-fidelity polymerase.

    • Design primers to include appropriate restriction sites for directional cloning into an expression vector (e.g., pET or pGEX systems).

    • Verify the sequence integrity of the cloned construct by DNA sequencing.

  • Protein Expression:

    • Transform the construct into an E. coli expression strain (BL21(DE3) or similar).

    • Induce protein expression using IPTG (typically 0.5-1 mM) at mid-log phase (OD600 0.6-0.8).

    • For optimal solubility, induce at lower temperatures (16-25°C) overnight rather than 37°C for shorter periods.

  • Protein Purification:

    • Lyse cells using sonication in a buffer containing 20 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, and protease inhibitors.

    • Purify using affinity chromatography:

      • For His-tagged constructs: Ni-NTA agarose with imidazole elution (gradient from 10 to 250 mM).

      • For GST-tagged constructs: Glutathione Sepharose with reduced glutathione elution.

    • Further purify by size exclusion chromatography to remove aggregates and ensure homogeneity.

    • Concentrate the protein and confirm purity by SDS-PAGE (should yield a single band at approximately 43 kDa) .

  • Quality Control:

    • Verify protein identity by Western blot using anti-EF-Tu antibodies.

    • Confirm protein activity by GTP binding assays if studying canonical EF-Tu function.

    • For surface binding studies, ensure the recombinant protein is properly folded using circular dichroism spectroscopy.

This purification strategy yields high-quality recombinant EF-Tu suitable for binding studies with host factors such as plasminogen and Factor H.

How can researchers analyze the interaction between EF-Tu and host plasma proteins?

Analyzing the interaction between Leptospira EF-Tu and host plasma proteins requires multiple complementary approaches:

  • Ligand Affinity Blotting:

    • Separate purified recombinant EF-Tu by SDS-PAGE under non-reducing conditions and transfer to nitrocellulose membranes.

    • Incubate membranes with diluted normal human serum (7% in PBS) as a source of plasma proteins for 90 minutes.

    • Wash thoroughly and probe with specific antibodies against the target plasma protein (e.g., goat anti-human FH at 1:10,000).

    • Detect binding using appropriate peroxidase-conjugated secondary antibodies and enhanced chemiluminescence .

  • ELISA-Based Binding Assays:

    • Coat microplate wells with purified recombinant EF-Tu (typically 1 μg/well).

    • Block non-specific binding sites with BSA or non-fat milk.

    • Add varying concentrations of purified plasma proteins or diluted serum.

    • Detect binding using specific antibodies against the target plasma protein.

    • For quantitative analysis, establish standard curves with known protein concentrations.

  • Surface Plasmon Resonance (SPR):

    • Immobilize recombinant EF-Tu on a sensor chip.

    • Flow solutions containing varying concentrations of plasma proteins over the chip.

    • Analyze binding kinetics (kon and koff rates) and calculate affinity constants (KD).

    • Perform competition assays to determine binding specificity.

  • Functional Assays:

    • For FH binding: Perform cofactor assays by incubating EF-Tu-bound FH with C3b and Factor I, then analyzing C3b cleavage fragments by immunoblotting .

    • For plasminogen binding: Incubate EF-Tu-bound plasminogen with plasminogen activator (e.g., uPA), then assess plasmin activity using chromogenic substrates or by fibrinogen degradation assays .

  • Binding Inhibition Studies:

    • To investigate the role of ionic interactions, perform binding assays in the presence of increasing NaCl concentrations.

    • To assess the involvement of lysine residues, include ε-aminocaproic acid in binding reactions .

These methods provide complementary data on binding specificity, affinity, and functional consequences of EF-Tu interactions with host plasma proteins.

What techniques are effective for detecting EF-Tu surface localization in Leptospira?

Multiple complementary techniques are recommended for convincingly demonstrating EF-Tu surface localization in Leptospira:

  • Immunofluorescence Microscopy:

    • Fix intact leptospires with 2% paraformaldehyde (avoid permeabilization).

    • Incubate with anti-EF-Tu antibodies followed by fluorophore-conjugated secondary antibodies.

    • As controls, include:

      • Known surface proteins (positive control)

      • Known cytoplasmic proteins (negative control)

      • Permeabilized samples (to confirm antibody reactivity)

    • Analyze using confocal microscopy to distinguish surface from internal staining.

  • Immunoelectron Microscopy:

    • Incubate intact leptospires with anti-EF-Tu antibodies and gold-conjugated secondary antibodies.

    • Analyze by transmission electron microscopy to visualize gold particles on the bacterial surface.

    • Quantify the distribution of gold particles between surface and internal locations.

  • Protease Accessibility Assays:

    • Treat intact leptospires with proteases that cannot penetrate the membrane (e.g., proteinase K).

    • Analyze EF-Tu degradation by immunoblotting compared to cytoplasmic protein controls.

    • A reduction in EF-Tu signal indicates surface exposure.

  • Surface Biotinylation:

    • Label intact bacteria with membrane-impermeable biotin reagents.

    • Lyse bacteria and isolate biotinylated proteins using streptavidin affinity chromatography.

    • Identify EF-Tu in the biotinylated fraction by immunoblotting.

    • Include controls for membrane integrity during biotinylation.

  • Flow Cytometry:

    • Incubate intact bacteria with anti-EF-Tu antibodies and fluorophore-conjugated secondary antibodies.

    • Analyze by flow cytometry to quantify surface binding.

    • Compare with permeabilized samples to establish the proportion of EF-Tu that is surface-accessible.

What methods are most reliable for differentiating between Leptospira interrogans serovars Icterohaemorrhagiae and Copenhageni?

Differentiating between the closely related Leptospira interrogans serovars Icterohaemorrhagiae and Copenhageni requires multiple complementary approaches:

  • Monoclonal Antibody (mAb) Typing:

    • The most reliable serological method uses a panel of specific mAbs:

      • F12C3: Agglutinates both Icterohaemorrhagiae and Copenhageni serovars

      • F70C14: Specific for Icterohaemorrhagiae serovar

      • F70C24: Specific for Copenhageni serovar

    • Agglutination patterns can definitively distinguish between these serovars, with titers typically around 10,000-30,000 .

  • Genetic Methods:

    • lic12008 Gene Sequence Analysis: All isolates identified as Icterohaemorrhagiae serovar have a characteristic single base insertion compared to Copenhageni serovar sequences .

    • Multiple-Locus Variable-Number Tandem Repeat Analysis (MLVA): This can reveal genetic diversity between isolates, with three loci showing differences in repeat numbers between strains .

  • Restriction Endonuclease Analysis (REA):

    • This method is NOT recommended for differentiation as it fails to discriminate between Icterohaemorrhagiae and Copenhageni serovars, yielding identical restriction patterns .

For definitive serovar determination, combining serological methods (particularly monoclonal antibody typing) with genetic analysis (especially lic12008 gene sequencing) provides the most reliable results. It's worth noting that while genetic tools reveal diversity within serovars, none of the genetic tools alone was able to definitively determine serovars in the study .

How does EF-Tu expression differ between pathogenic and non-pathogenic Leptospira species?

EF-Tu expression appears to be consistent across both pathogenic and non-pathogenic Leptospira species based on current evidence:

  • Protein Expression Levels:

    • Immunoblot analysis using anti-EF-Tu serum detected the 43 kDa EF-Tu protein in all tested Leptospira strains, including both pathogenic serovars (Panama, Javanica, Tarassovi, Cynopteri, Copenhageni, Pomona, and Shermani) and the non-pathogenic saprophytic L. biflexa serovar Patoc .

    • The intensity of detection bands in immunoblot analyses suggests similar expression levels across strains, though quantitative proteomic studies would be needed to confirm this.

  • Sequence Conservation:

    • EF-Tu from pathogenic Leptospira strains is highly conserved (99-100% identity at the amino acid level).

    • L. interrogans Copenhageni L1-130 and L. biflexa Patoc1 EF-Tu share 86% sequence identity .

  • Functional Differences:

    • While both pathogenic and non-pathogenic strains express EF-Tu, its contribution to virulence in pathogenic strains comes from its moonlighting activities when surface-exposed.

    • The presence of EF-Tu in non-pathogenic bacteria does not confer survival advantages in hosts because pathogenic strains have additional virulence factors not present in saprophytic species .

It's important to note that the key distinction lies not in the expression level of EF-Tu itself, but in how pathogenic Leptospira employ this protein as part of a broader virulence strategy. Pathogenic species possess multiple additional virulence factors absent in non-pathogenic species, including other complement regulator-binding proteins like LenA, LenB, LigA, LigB, and LcpA .

How does the virulence of Leptospira interrogans serovar Copenhageni compare to other serovars in clinical settings?

Leptospira interrogans serovar Copenhageni is among the most virulent serovars in clinical settings:

  • Prevalence in Severe Human Infections:

    • Strains belonging to serovars Icterohaemorrhagiae and Copenhageni are considered the most pathogenic Leptospira serovars.

    • Together, they represent more than half of the leptospires encountered in severe human infections globally .

    • In urban areas, particularly in South American cities like the favelas of Brazil, the majority of acute leptospirosis cases are caused by serovar Copenhageni .

  • Geographic Distribution:

    • In the United Kingdom, Copenhageni is the predominant serovar in the Icterohaemorrhagiae serogroup, accounting for 84.2% (16/19) of isolates examined in one study .

    • This predominance is likely reflected in other regions where rats serve as maintenance hosts.

  • Disease Severity:

    • Infections with serovar Copenhageni are frequently associated with severe manifestations of leptospirosis, including Weil's disease, characterized by jaundice, renal failure, and hemorrhagic manifestations.

    • The terms "Weil's disease" and "Icterohaemorrhagiae infection" have become synonymous with the acute icteric form of leptospirosis transmitted by brown rats (Rattus norvegicus) .

  • Subclinical Infections:

    • Despite its virulence, research suggests that many infections with Leptospira (including Copenhageni) may be subclinical or mild.

    • Studies indicate there may be as many as 191 infections for every one clinical case of disease .

The high virulence of serovar Copenhageni is likely related to its multiple immune evasion strategies, including the surface expression of EF-Tu which contributes to complement resistance and tissue invasion . This makes Copenhageni infections particularly concerning in clinical settings, especially in populations with occupational or environmental exposure to rat reservoirs.

What is the role of EF-Tu in the pathogenesis of severe leptospirosis manifestations?

EF-Tu contributes to severe leptospirosis manifestations through multiple pathogenic mechanisms:

  • Complement Evasion and Persistent Infection:

    • EF-Tu binds Factor H and plasminogen, leading to complement inactivation through C3b degradation .

    • This immune evasion strategy allows Leptospira to avoid clearance, leading to higher bacterial loads and disseminated infection.

    • Persistent infection contributes to prolonged inflammatory responses and tissue damage characteristic of severe leptospirosis.

  • Enhanced Tissue Invasion:

    • EF-Tu mediates binding to extracellular matrix components (collagen I and IV, fibronectin, laminin, elastin) .

    • Plasminogen bound to EF-Tu is converted to active plasmin, which degrades tissue barriers.

    • These mechanisms facilitate bacterial dissemination to multiple organs, including liver and kidneys, which are primary targets in severe leptospirosis.

  • Coagulation System Interaction:

    • EF-Tu binds fibrinogen directly and can degrade it through EF-Tu-bound plasmin .

    • This interaction with the coagulation system may contribute to the hemorrhagic manifestations observed in severe cases of leptospirosis.

    • Fibrinogen degradation products can also have pro-inflammatory effects, potentially exacerbating tissue damage.

  • Synergy with Other Virulence Factors:

    • EF-Tu works in concert with other leptospiral virulence factors.

    • Pathogenic Leptospira possess multiple complement evasion strategies, including other surface proteins like LenA, LenB, LigA, LigB, and LcpA .

    • They also secrete proteases that cleave key complement proteins.

    • This multi-faceted approach to immune evasion contributes to the severity of infection.

Understanding EF-Tu's contribution to pathogenesis is important, but it's crucial to recognize that severe leptospirosis results from the collective action of multiple virulence factors. The presence of EF-Tu alone is insufficient for virulence, as evidenced by its presence in non-pathogenic Leptospira species .

What are the optimal experimental models for studying Leptospira EF-Tu in the context of host-pathogen interactions?

Several experimental models offer distinct advantages for studying Leptospira EF-Tu in host-pathogen interactions:

  • In Vitro Models:

    • Cell Culture Systems:

      • Human endothelial cells for studying vascular interactions

      • Kidney epithelial cells for modeling renal colonization

      • Macrophages for immune response studies

    • Ex Vivo Tissue Models:

      • Perfused liver or kidney slices to study tissue tropism

      • Whole blood assays for complement and coagulation studies

    • Methodology: Use fluorescently-labeled recombinant EF-Tu or EF-Tu-expressing Leptospira, with isogenic EF-Tu mutants as controls.

  • Animal Models:

    • Hamsters:

      • Golden Syrian hamsters are highly susceptible to leptospirosis

      • Develop acute disease similar to severe human leptospirosis

      • Previously used for evaluating EF-Tu as a vaccine candidate

    • Guinea Pigs:

      • Develop symptoms similar to human leptospirosis

      • Useful for studying kidney colonization

    • Rats:

      • Natural reservoir for Icterohaemorrhagiae and Copenhageni serovars

      • Valuable for studying chronic carrier states and transmission

    • Methodology: Compare wild-type Leptospira with strains engineered to overexpress or underexpress EF-Tu, or with modified EF-Tu binding domains.

  • Comparative Models:

    • Multi-Species Comparison:

      • Compare EF-Tu function between pathogenic and non-pathogenic Leptospira

      • Examine moonlighting activities across different bacterial genera

    • Multi-Serovar Comparison:

      • Compare EF-Tu functionality between Copenhageni and other serovars

      • Correlate with clinical virulence patterns

    • Methodology: Employ protein domain swapping or site-directed mutagenesis to identify functional regions.

  • Molecular Interaction Models:

    • Protein-Protein Interaction Assays:

      • Surface plasmon resonance for binding kinetics

      • Yeast two-hybrid screens to identify novel host interaction partners

    • Structural Biology Approaches:

      • X-ray crystallography of EF-Tu complexed with host factors

      • Hydrogen-deuterium exchange mass spectrometry to map binding interfaces

    • Methodology: Generate structure-based hypotheses for targeted mutagenesis.

The optimal approach combines multiple models, starting with molecular and in vitro studies to characterize specific interactions, followed by animal models to validate physiological relevance. Including both acute disease models (hamsters) and reservoir host models (rats) provides complementary insights into EF-Tu's role in both pathogenesis and persistent infection.

What are promising strategies for targeting EF-Tu to develop novel therapeutics against leptospirosis?

Several promising strategies could leverage our understanding of EF-Tu to develop novel therapeutics against leptospirosis:

  • Inhibition of EF-Tu Moonlighting Functions:

    • Develop small molecule inhibitors or peptide mimetics that specifically block EF-Tu interactions with host molecules without affecting protein synthesis.

    • Target the binding sites for plasminogen and Factor H on the EF-Tu molecule to prevent immune evasion .

    • This approach would impair bacterial dissemination and complement evasion while minimizing selection pressure, as the primary function of EF-Tu would remain intact.

  • Combination Therapies Targeting Multiple Moonlighting Proteins:

    • Develop cocktails of inhibitors targeting multiple moonlighting proteins simultaneously (EF-Tu, LenA, LenB, LigA, LigB, LcpA) .

    • This multi-target approach could overcome the redundancy in virulence mechanisms and reduce the likelihood of resistance development.

  • Structure-Based Drug Design:

    • Utilize structural information about EF-Tu's moonlighting binding sites to design highly specific inhibitors.

    • Focus on regions of EF-Tu that are unique to Leptospira to enhance specificity and reduce effects on host protein synthesis machinery.

  • Attenuated Vaccine Development:

    • Engineer Leptospira strains with modified EF-Tu that maintains protein synthesis function but lacks moonlighting activities.

    • Such strains could serve as live attenuated vaccines with reduced virulence but preserved immunogenicity.

  • Epitope-Based Vaccine Improvement:

    • Although EF-Tu alone was not protective as a vaccine candidate , combining specific EF-Tu epitopes with other protective antigens might enhance vaccine efficacy.

    • Focus on epitopes of EF-Tu involved in moonlighting functions rather than the whole protein.

  • Anti-Virulence Approach:

    • Develop compounds that prevent surface localization of EF-Tu without affecting its expression.

    • This would specifically target the pathogenic role of EF-Tu while avoiding selection pressure on its essential function.

These strategies represent a shift from traditional antibiotic approaches toward targeting specific virulence mechanisms, potentially leading to therapeutics with fewer side effects and reduced pressure for resistance development. Advancing these approaches requires further research into the structural basis of EF-Tu moonlighting functions and their relative importance in different stages of infection.

How might comparative genomics and proteomics advance our understanding of EF-Tu evolution in Leptospira?

Comparative genomics and proteomics offer powerful approaches to understand EF-Tu evolution in Leptospira:

  • Evolutionary Trajectory Analysis:

    • Sequence comparison across all known Leptospira species (currently >60 species) could reveal the evolutionary history of EF-Tu.

    • Phylogenetic analysis can identify when moonlighting functions were acquired or enhanced during Leptospira evolution.

    • Comparison of selection pressures on different domains could distinguish regions involved in protein synthesis versus moonlighting functions.

  • Structure-Function Correlation:

    • Mapping sequence variations onto 3D structures can identify critical regions for moonlighting functions.

    • Comparative structural prediction across pathogenic and non-pathogenic species may reveal subtle conformational differences affecting surface exposure.

    • This could explain why EF-Tu in non-pathogenic species doesn't confer the same virulence advantages despite high sequence similarity (86%) .

  • Post-Translational Modification (PTM) Analysis:

    • Comparative proteomics could identify PTMs unique to pathogenic Leptospira EF-Tu.

    • Such modifications might influence protein localization, stability, or host interaction capabilities.

    • Mass spectrometry techniques could map these modifications and their distribution across different Leptospira species.

  • Regulome Analysis:

    • Comparing transcriptional and translational regulation of EF-Tu across species and environmental conditions.

    • Identifying regulatory elements that differ between pathogenic and non-pathogenic species.

    • Understanding how environmental signals modulate EF-Tu expression and surface localization.

  • Horizontal Gene Transfer Assessment:

    • Analysis of codon usage and genomic context to identify potential horizontal gene transfer events.

    • Comparing Leptospira EF-Tu with that of other bacteria known to share ecological niches.

    • This could reveal if moonlighting capabilities were acquired through horizontal gene transfer from other bacterial species.

  • Pangenome Analysis:

    • Examining the full genetic repertoire across all Leptospira species to contextualize EF-Tu within the broader evolution of virulence.

    • Identifying genes consistently co-occurring with specific EF-Tu variants that might functionally interact with or complement EF-Tu's moonlighting activities.

These approaches would provide a comprehensive evolutionary perspective on how EF-Tu has been adapted in Leptospira, potentially identifying key transitions that enabled its expansion from a housekeeping protein to a multifunctional virulence factor. This evolutionary understanding could inform both fundamental bacteriology and applied therapeutic development.

What are the key considerations for ensuring reproducibility in studies of recombinant EF-Tu function?

Ensuring reproducibility in studies of recombinant Leptospira EF-Tu function requires attention to several critical factors:

  • Protein Expression and Purification Standardization:

    • Consistently use the same expression system and host strain.

    • Document and maintain consistent induction conditions (temperature, duration, inducer concentration).

    • Implement standardized purification protocols with defined buffer compositions.

    • Include quality control steps: SDS-PAGE, Western blot, and circular dichroism to confirm protein integrity and folding.

    • Quantify endotoxin levels, as contamination can affect functional assays.

  • Host Component Standardization:

    • For studies using human serum as a source of FH or plasminogen, establish serum pools from multiple donors to minimize individual variation.

    • Consider using purified components with defined concentrations for binding studies.

    • Document the source, purification method, and quality control tests for all host proteins.

    • Test multiple lots of commercial proteins to ensure consistent activity.

  • Assay Validation and Controls:

    • Include positive and negative controls in every experiment (e.g., LigBC and LIC10301 for FH binding studies) .

    • Establish dose-response relationships rather than single-point measurements.

    • Use multiple complementary methods to confirm interactions (e.g., ELISA, surface plasmon resonance, ligand blotting).

    • Develop quantitative readouts with clearly defined thresholds for positivity.

  • Detailed Methodology Reporting:

    • Provide complete buffer compositions, including pH and additives.

    • Report protein concentrations, incubation times, temperatures, and washing conditions.

    • Specify the exact region of the tuf gene used for recombinant protein production.

    • Include detailed statistical methods for data analysis.

  • Addressing Batch-to-Batch Variation:

    • Prepare large batches of recombinant protein and aliquot for long-term studies.

    • Implement reference standards to normalize results across batches.

    • Regularly test protein activity against standard assays.

    • Document and report batch numbers in publications.

  • Physiological Relevance Considerations:

    • Perform assays under conditions mimicking the host environment (pH, ionic strength, temperature).

    • Include whole-cell validation experiments with intact Leptospira to confirm relevance of findings with recombinant protein.

    • Compare results across multiple Leptospira strains to ensure generalizability.

By systematically addressing these factors, researchers can significantly enhance the reproducibility of studies on Leptospira EF-Tu function, enabling more reliable cross-laboratory comparisons and accelerating progress in understanding this moonlighting protein's role in leptospiral pathogenesis.

How can researchers effectively differentiate between EF-Tu's canonical and moonlighting functions in experimental settings?

Differentiating between EF-Tu's canonical protein synthesis function and its moonlighting activities requires sophisticated experimental approaches:

  • Genetic Manipulation Strategies:

    • Domain-Specific Mutations: Generate EF-Tu variants with mutations in domains predicted to be involved in moonlighting functions but not protein synthesis.

    • Conditional Expression Systems: Develop strains with tunable EF-Tu expression to identify threshold levels needed for different functions.

    • Heterologous Expression: Express Leptospira EF-Tu in non-pathogenic bacteria to assess which functions transfer independently of other leptospiral proteins.

  • Subcellular Localization Studies:

    • Fractionation Analysis: Quantitatively compare cytoplasmic versus surface EF-Tu pools using carefully controlled fractionation procedures.

    • Dual-Function Tracking: Develop dual-reporter systems to simultaneously track protein synthesis activity and surface localization.

    • Super-Resolution Microscopy: Visualize the precise subcellular distribution of EF-Tu during different growth phases.

  • Functional Uncoupling Approaches:

    • Competitive Inhibition: Use GTP analogs that block protein synthesis function without affecting surface-exposed EF-Tu.

    • Surface-Specific Targeting: Apply membrane-impermeable labeling or inhibition techniques that only affect surface-exposed EF-Tu.

    • Temperature-Sensitive Mutants: Generate conditional mutants that selectively lose one function at restrictive temperatures.

  • Complementation Studies:

    • Function-Specific Complementation: In EF-Tu-depleted backgrounds, express variants that can restore only protein synthesis or only moonlighting functions.

    • Heterologous Complementation: Test whether EF-Tu from non-pathogenic species can restore moonlighting functions in pathogenic strains.

  • Temporal Regulation Analysis:

    • Time-Course Studies: Track the allocation of EF-Tu between translation and moonlighting functions during different growth phases.

    • Stress Response Analysis: Examine how various stressors shift the balance between canonical and moonlighting functions.

  • Structure-Based Approaches:

    • Conformational Analysis: Investigate whether EF-Tu undergoes structural changes when transitioning between functions.

    • Binding Site Mapping: Use hydrogen-deuterium exchange mass spectrometry to identify regions involved in moonlighting interactions.

    • Crosslinking Studies: Apply chemical crosslinking to capture EF-Tu interacting with components of the translation machinery versus host factors.

These approaches, especially when used in combination, allow researchers to dissect the multifunctional nature of EF-Tu and understand how a single protein can participate in such diverse cellular processes. The insights gained could inform strategies for selectively targeting moonlighting functions without disrupting essential protein synthesis, potentially leading to novel therapeutic approaches with reduced side effects.

What are the most significant unanswered questions regarding Leptospira EF-Tu that should direct future research?

Several critical questions about Leptospira EF-Tu remain unanswered and should guide future research:

  • Mechanism of Surface Localization:

    • How is EF-Tu, traditionally a cytoplasmic protein, transported to the bacterial surface?

    • Does this process involve specific secretion systems, membrane transporters, or cell lysis?

    • Is surface localization regulated in response to environmental conditions or host factors?

  • Structural Basis of Moonlighting Functions:

    • What structural features of EF-Tu enable its diverse binding capabilities?

    • Does surface-localized EF-Tu adopt a different conformation than cytoplasmic EF-Tu?

    • Which specific domains or residues mediate interactions with different host factors?

  • Functional Hierarchy in Pathogenesis:

    • What is the relative contribution of EF-Tu to virulence compared to other leptospiral moonlighting proteins?

    • In which stages of infection (adhesion, invasion, immune evasion, persistence) is EF-Tu most crucial?

    • How does its contribution vary across different host species and tissues?

  • Regulatory Networks:

    • How is the expression and surface localization of EF-Tu regulated during infection?

    • Do environmental signals in different host niches modulate EF-Tu function?

    • Is there coordination between EF-Tu and other virulence factors?

  • Host Response Interactions:

    • Beyond complement and coagulation systems, does EF-Tu interact with other host defense mechanisms?

    • Does EF-Tu trigger specific innate immune recognition pathways?

    • Can adaptive immunity effectively target surface-exposed EF-Tu despite its high conservation?

  • Therapeutic Targeting Potential:

    • Can EF-Tu moonlighting functions be selectively inhibited without affecting bacterial viability?

    • Would such inhibition significantly attenuate virulence in vivo?

    • Can structural knowledge of EF-Tu be leveraged for rational drug design?

  • Evolutionary Significance:

    • When did EF-Tu acquire moonlighting capabilities during Leptospira evolution?

    • Why do non-pathogenic species maintain EF-Tu with high sequence similarity despite lacking virulence?

    • Has host-pathogen co-evolution shaped EF-Tu functionality?

  • Cross-Species Relevance:

    • How do EF-Tu functions in Leptospira compare with those in other bacterial pathogens?

    • Could insights from Leptospira EF-Tu inform understanding of similar proteins in other species?

    • Are there common mechanisms that could be targeted for broad-spectrum therapeutic development?

Addressing these questions will require integrated approaches combining structural biology, molecular genetics, advanced imaging, and in vivo models. The answers would significantly advance our understanding of leptospiral pathogenesis and potentially reveal new strategies for intervention against this important zoonotic disease.

How might technological advances in structural biology and systems biology transform our understanding of EF-Tu's multifunctional nature?

Emerging technologies in structural and systems biology offer transformative potential for understanding EF-Tu's multifunctional nature:

  • Cryo-Electron Microscopy Advances:

    • Single-particle cryo-EM can now achieve near-atomic resolution of proteins in different conformational states.

    • This could reveal how EF-Tu structurally transitions between its canonical role in translation and its moonlighting functions.

    • Cryo-electron tomography could visualize EF-Tu in its native context on the leptospiral surface, providing insights into its spatial organization and interactions.

  • Integrative Structural Biology:

    • Combining X-ray crystallography, NMR, SAXS, and computational modeling to build complete models of EF-Tu complexes with various host factors.

    • Hydrogen-deuterium exchange mass spectrometry can map binding interfaces under near-physiological conditions.

    • These approaches could identify structural adaptations that enable EF-Tu's multifunctionality.

  • Single-Molecule Techniques:

    • Single-molecule FRET to track conformational changes in EF-Tu when interacting with different binding partners.

    • Optical tweezers to measure binding forces between EF-Tu and host molecules.

    • These techniques could reveal dynamic aspects of EF-Tu function impossible to observe in bulk assays.

  • Advanced Proteomics:

    • Proximity labeling techniques (BioID, APEX) to identify the complete interactome of EF-Tu in living bacteria.

    • Quantitative proteomics to track changes in EF-Tu associations during different phases of infection.

    • Crosslinking mass spectrometry to capture transient interactions in native conditions.

  • Systems Biology Integration:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics to place EF-Tu within broader regulatory networks.

    • Mathematical modeling of EF-Tu's dual functions to understand how resources are allocated between protein synthesis and moonlighting.

    • Network analysis to identify hub proteins that coordinate with EF-Tu during host interaction.

  • In Situ Structural Biology:

    • Correlative light and electron microscopy to visualize EF-Tu during host-pathogen interactions at nanometer resolution.

    • Cryo-focused ion beam milling combined with tomography to observe EF-Tu within intact host-pathogen interfaces.

    • These approaches could reveal the structural basis of EF-Tu function in its native context.

  • AlphaFold and Deep Learning Applications:

    • AI-driven structure prediction to model EF-Tu interactions with various host factors.

    • Deep learning approaches to predict functional consequences of EF-Tu sequence variations across Leptospira species.

    • These computational tools could accelerate hypothesis generation and experimental design.

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.