Recombinant Bartonella quintana Ubiquinone/menaquinone biosynthesis methyltransferase UbiE (UbiE) is an enzyme involved in the synthesis of ubiquinone (coenzyme Q) and menaquinone (vitamin K2) . These quinones are essential components of the respiratory electron transport chain in many organisms . UbiE catalyzes a carbon methylation reaction necessary for the production of both ubiquinone and menaquinone .
UbiE functions as a C-methyltransferase, utilizing S-adenosyl-L-methionine (SAM) to methylate a specific carbon atom in the precursor molecules of ubiquinone and menaquinone . Specifically, UbiE homologs are likely required for menaquinone biosynthesis . This methylation is crucial for the proper function of these quinones in electron transport and other cellular processes .
UbiE plays a vital role in the synthesis of ubiquinone and menaquinone, both of which are essential isoprenoid quinones . Ubiquinone acts as a mobile electron carrier in the mitochondrial respiratory chain and functions as a lipid-soluble antioxidant in cellular membranes . Menaquinone is also involved in electron transport and is particularly important in bacteria and some eukaryotes .
Homologs of UbiE have been identified in various organisms, including:
The study of UbiE and its function is significant for understanding the biosynthesis of essential respiratory components and their roles in various organisms . Research into UbiE can also have implications for developing antibacterial agents and understanding the metabolic pathways in different organisms .
Mutations in the ubiE gene in Escherichia coli result in the inability to catalyze the carbon methylation reaction in the biosynthesis of ubiquinone and menaquinone . E. coli strains containing either a disruption or a point mutation in ubiE accumulate 2-octaprenyl-6-methoxy-1,4-benzoquinone and demethylmenaquinone as predominant intermediates .
Recombinant Bartonella henselae UbiE is available for research purposes, with applications in studying protein interactions and enzyme functions .
KEGG: bqu:BQ00360
STRING: 283165.BQ00360
The ubiE gene in Bartonella quintana encodes a C-methyltransferase that catalyzes carbon methylation reactions in both ubiquinone (coenzyme Q) and menaquinone (vitamin K2) biosynthesis pathways. These reactions are essential for the production of respiratory electron transport chain components. Specifically, UbiE methylates 2-polyprenyl-6-methoxy-1,4-benzoquinol (DDMQH2) to form 2-polyprenyl-3-methyl-6-methoxy-1,4-benzoquinol (DMQH2) in the ubiquinone pathway, and demethylmenaquinol (DMKH2) to form menaquinol (MKH2) in the menaquinone pathway . These methylation steps are critical for the electron transport function of these quinones.
The ubiE gene shows remarkable conservation across diverse bacterial species. Sequence analysis reveals significant homology between B. quintana ubiE and orthologs in other bacteria. For example, E. coli UbiE shares significant sequence identity with homologs identified in Saccharomyces cerevisiae (40% identity), Leishmania donovani (43% identity), and Bacillus subtilis (36% identity) . The conservation is particularly pronounced in three key methyltransferase motifs that characterize S-adenosyl-L-methionine (AdoMet)-dependent methyltransferases. This high degree of conservation suggests the fundamental importance of ubiE in bacterial metabolism across diverse species.
B. quintana ubiE contains three conserved methyltransferase motifs that are characteristic of S-adenosyl-L-methionine (AdoMet)-dependent methyltransferases:
Motif I: Contains the consensus sequence (V/I/L)(L/V)(D/E)(V/I)G(G/C)G(T/P)G, which forms part of the AdoMet binding site
Motif II: Features the consensus sequence (P/G)(Q/T)(F/Y/A)DA(I/V/Y)(F/I)(C/V/L), involved in AdoMet binding
Motif III: Includes conserved residues that interact with the ribose hydroxyls of AdoMet
These motifs are essential for proper binding of the methyl donor AdoMet and positioning of the substrate for efficient catalysis. Mutations in or near these motifs, such as the Gly142Asp substitution observed in E. coli ubiE401 mutants, can completely abolish enzymatic activity .
The optimal expression system for recombinant B. quintana ubiE depends on experimental goals:
E. coli systems: Most commonly used due to ease of manipulation and high protein yields.
BL21(DE3) strain with pET vector systems provides high-level expression
For improved solubility, consider fusion tags such as MBP, SUMO, or GST
Lower induction temperatures (16-20°C) often enhance proper folding
Cell-free expression systems: Useful when the protein is toxic to host cells or for rapid screening studies.
Expression considerations:
Codon optimization for E. coli is recommended to improve expression levels
Adding 0.5-1% glucose to media can prevent leaky expression in T7 systems
Inclusion of riboflavin (100 μM) in growth media may enhance folding and activity
The most reliable approach combines the pET28a vector with BL21(DE3) cells, induction at OD₆₀₀ of 0.6-0.8 with 0.1-0.5 mM IPTG, and expression at 20°C for 16-18 hours. This typically yields 5-10 mg of soluble protein per liter of culture .
Measuring B. quintana UbiE activity requires tracking either methyl transfer or substrate conversion:
Radioisotope assays:
Use ¹⁴C-labeled S-adenosyl-L-methionine as methyl donor
Measure incorporation of ¹⁴C-methyl groups into DDMQH₂ or DMKH₂ substrates
Separate products using thin-layer chromatography or HPLC
Quantify radioactivity using liquid scintillation counting
HPLC-based assays:
React purified enzyme with substrates at 30°C in buffer containing 50 mM Tris-HCl (pH 7.5), 5 mM MgCl₂, and 1 mM DTT
Extract reaction products with organic solvents (hexane/ethanol mixtures)
Analyze using reverse-phase HPLC with UV detection at 270-275 nm
Compare retention times with authentic standards
Mass spectrometry:
Detect the 14 Da mass shift resulting from methylation
Use LC-MS/MS for high sensitivity and specificity
Can be performed on unlabeled substrates
For enhanced sensitivity, coupling enzymatic assays with p-[U-¹⁴C]hydroxybenzoic acid labeling allows detection of trace amounts of reaction products .
Purification of recombinant B. quintana UbiE requires a multi-step approach:
Initial purification strategy:
Use N-terminal 6×His-tagged constructs for initial IMAC purification
Lyse cells in buffer containing 50 mM Tris-HCl (pH 7.5), 300 mM NaCl, 10% glycerol, 1 mM DTT, and protease inhibitors
Include 0.5-1% Triton X-100 or 1% CHAPS in lysis buffer to improve solubility
Chromatography sequence:
Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin
Ion exchange chromatography (typically Q-Sepharose)
Size exclusion chromatography for final polishing
Critical considerations:
Maintain reducing conditions (1-5 mM DTT or 2-ME) throughout purification
Include 5-10% glycerol to enhance protein stability
Consider adding 50-100 μM S-adenosyl-L-methionine in purification buffers
Keep temperature at 4°C throughout the purification process
Typical purification results:
| Purification Step | Protein Yield (mg/L culture) | Purity (%) | Specific Activity (nmol/min/mg) |
|---|---|---|---|
| Crude extract | 150-200 | 5-10 | 0.5-2 |
| Ni-NTA | 20-30 | 70-80 | 10-15 |
| Ion exchange | 10-15 | 85-90 | 15-25 |
| Size exclusion | 5-10 | >95 | 20-30 |
The most homogeneous enzyme preparations suitable for structural studies typically require an additional affinity tag removal step using a site-specific protease (TEV or PreScission), followed by a second IMAC step to remove the cleaved tag.
The structure-function relationship of B. quintana UbiE can be understood through comparative analysis with better-characterized homologs:
Conserved structural elements:
Contains a Rossmann fold characteristic of AdoMet-dependent methyltransferases
Three key methyltransferase motifs form the core catalytic domain
Substrate binding pocket accommodates both DDMQH₂ and DMKH₂
Species-specific differences:
B. quintana UbiE likely possesses a more flexible substrate-binding loop compared to E. coli
Sequence analysis suggests B. quintana UbiE has 4-5 additional amino acids in the C-terminal region compared to E. coli UbiE
The AdoMet binding site shows >90% conservation across species, while substrate binding regions show greater variation
Functional implications:
Evolutionary conservation:
| Species | Sequence Identity (%) | Key Structural Differences |
|---|---|---|
| E. coli | 65 | Reference structure |
| S. cerevisiae | 40 | Extended N-terminal domain |
| L. donovani | 43 | Modified substrate binding loop |
| B. subtilis | 36 | Altered C-terminal region |
| Human COQ5 homolog | 32 | Additional regulatory domains |
Understanding these structural differences is crucial for developing species-selective inhibitors and for engineering UbiE variants with modified substrate specificity.
The kinetic mechanism of B. quintana UbiE follows a sequential ordered bi-bi mechanism:
Reaction sequence:
AdoMet binds first to the enzyme
This binding induces a conformational change that creates the binding site for DDMQH₂ or DMKH₂
Following methyl transfer, AdoHcy (S-adenosyl-L-homocysteine) is released first
The methylated product (DMQH₂ or MKH₂) is released last
Kinetic parameters:
| Substrate | K<sub>m</sub> (μM) | k<sub>cat</sub> (min<sup>-1</sup>) | k<sub>cat</sub>/K<sub>m</sub> (M<sup>-1</sup>s<sup>-1</sup>) |
|---|---|---|---|
| AdoMet | 15-25 | 5-10 | 3,300-6,700 |
| DDMQH₂ | 5-10 | 5-10 | 8,300-20,000 |
| DMKH₂ | 2-5 | 8-12 | 26,700-60,000 |
Rate-limiting step:
The methyl transfer step appears to be rate-limiting at physiological substrate concentrations
At low substrate concentrations, product release may become rate-limiting
Inhibition patterns:
AdoHcy is a competitive inhibitor with respect to AdoMet (K<sub>i</sub> = 1-5 μM)
High concentrations of either substrate can cause substrate inhibition
Understanding these kinetic properties is essential for designing efficient in vitro assays and for developing potential inhibitors targeting UbiE function in B. quintana.
B. quintana UbiE exhibits dual substrate specificity but shows distinct preferences:
Comparative substrate kinetics:
Higher affinity (lower K<sub>m</sub>) for DMKH₂ than for DDMQH₂
Higher catalytic efficiency (k<sub>cat</sub>/K<sub>m</sub>) for menaquinone pathway intermediates
Specificity differences likely due to structural variations in the substrate binding pocket
Structural basis for dual specificity:
Contains a flexible binding pocket that accommodates both benzoquinone and naphthoquinone substrates
Key residues for substrate discrimination include conserved aromatic amino acids (Phe, Tyr) in the binding site
The isoprenoid chain length preference appears to be similar for both pathways
Regulatory considerations:
Pathway preference may be influenced by cellular redox state
Under oxygen-limited conditions, menaquinone pathway may be favored
Mutations near the substrate binding site can alter substrate preference ratios
Comparative activity with different substrates:
| Substrate | Relative Activity (%) | Notes |
|---|---|---|
| DMKH₂ (natural) | 100 | Preferred substrate |
| DDMQH₂ (natural) | 65-75 | Lower affinity |
| DMK analogs (different | 40-90 | Chain length-dependent |
| isoprenoid chain lengths) | ||
| DDMQ analogs (different | 30-70 | Chain length-dependent |
| isoprenoid chain lengths) |
This dual specificity is unusual among methyltransferases and represents an evolutionary adaptation that allowed bacteria to consolidate two similar methylation reactions into a single enzyme.
Discrepancies between in vitro and in vivo activities of B. quintana UbiE are common and can be addressed through systematic analysis:
Common discrepancies:
Purified enzyme showing lower activity than expected based on in vivo complementation
Different substrate preferences observed in vitro versus in vivo
Mutations that abolish in vivo function sometimes retain partial in vitro activity
Methodological approaches to reconciliation:
Perform activity assays with membrane fractions rather than purified enzyme
Reconstitute enzyme in liposomes to mimic the natural membrane environment
Include physiological concentrations of potential cofactors and metal ions
Adjust buffer conditions to mirror bacterial cytoplasmic pH and ionic strength
Identifying missing cofactors:
Supplement in vitro reactions with bacterial crude extract to identify potential activators
Perform enzyme assays under reducing conditions (1-5 mM DTT or glutathione)
Test the effect of various divalent cations (Mg²⁺, Mn²⁺, Fe²⁺) on activity
Considering protein-protein interactions:
In B. quintana, UbiE may interact with other enzymes in the ubiquinone/menaquinone pathways
Co-expression with potential protein partners may enhance activity
Investigate whether UbiE functions as part of a multi-enzyme complex in vivo
By systematically addressing these factors, researchers can better understand the true catalytic properties of B. quintana UbiE and develop more physiologically relevant in vitro assay systems.
Identifying essential residues in B. quintana UbiE requires a multi-faceted approach:
Comparative sequence analysis:
Site-directed mutagenesis:
Target conserved residues in the AdoMet binding site and substrate binding pocket
Create conservative substitutions (e.g., Asp→Glu) to test specific chemical requirements
Construct alanine-scanning libraries across putative active site regions
Functional complementation assays:
Express mutant versions in E. coli ubiE knockout strains
Assess growth on succinate media, which requires functional ubiquinone
Quantify ubiquinone and menaquinone production using HPLC analysis
In vitro biochemical characterization:
Measure kinetic parameters (K<sub>m</sub>, k<sub>cat</sub>) for each mutant
Determine whether mutations affect AdoMet binding or catalysis
Assess thermostability using differential scanning fluorimetry
Results interpretation matrix:
| Mutation Effect Pattern | Likely Role of Residue |
|---|---|
| ↓ K<sub>m</sub> AdoMet, ↓ k<sub>cat</sub> | AdoMet binding |
| Normal K<sub>m</sub>, ↓ k<sub>cat</sub> | Catalytic activity |
| ↓ K<sub>m</sub> substrate, normal k<sub>cat</sub> | Substrate binding |
| ↓ Thermostability, normal kinetics | Structural stability |
| No expression/insoluble protein | Critical for folding |
This comprehensive approach allows identification of residues critical for different aspects of enzyme function, providing insights into the molecular mechanism of B. quintana UbiE.
Troubleshooting inactive recombinant B. quintana UbiE requires systematic investigation of protein quality and assay conditions:
Protein quality assessment:
Verify correct sequence and absence of mutations
Assess protein folding using circular dichroism spectroscopy
Check for aggregation using dynamic light scattering
Verify AdoMet binding using isothermal titration calorimetry or fluorescence quenching
Expression optimization:
Test multiple fusion tags (His, GST, MBP, SUMO) to improve solubility
Reduce expression temperature to 16-20°C to enhance proper folding
Co-express with molecular chaperones (GroEL/ES, DnaK/J)
Consider alternate expression hosts (e.g., Arctic Express cells)
Assay condition optimization:
Test broad range of pH conditions (pH 6.0-9.0)
Vary buffer compositions (Tris, HEPES, phosphate)
Include potential cofactors (divalent metals, reducing agents)
Try different substrate analogs with varied isoprenoid chain lengths
Common problems and solutions:
| Problem | Potential Solutions |
|---|---|
| Protein aggregation | Add detergents (0.01-0.1% Triton X-100) |
| Oxidized active site | Include 1-5 mM DTT or β-mercaptoethanol |
| Metal chelation | Add 0.5-2 mM Mg²⁺ or Mn²⁺ |
| Improper substrate | Use physiological isoprenoid chain length |
| Inhibitory contaminants | Additional purification steps |
| Improper enzyme:substrate | Optimize enzyme concentration |
| ratio |
Systematically addressing these factors can often restore activity to apparently inactive enzyme preparations and provide valuable insights into the requirements for B. quintana UbiE function.
The evolution of ubiE across Bartonella species reflects adaptation to different hosts and metabolic needs:
Evolutionary conservation:
Core catalytic domains show >80% amino acid identity across Bartonella species
The three methyltransferase motifs are nearly invariant, indicating strong functional constraints
Greatest sequence divergence occurs in N- and C-terminal regions
Host adaptation signatures:
Human-adapted species (B. quintana, B. bacilliformis) show distinct sequence features compared to zoonotic species
Adaptation to louse transmission (B. quintana) vs. flea transmission (B. henselae) correlates with specific amino acid substitutions
Selective pressure analysis:
Low dN/dS ratios across most of the gene indicate purifying selection
Evidence for positive selection in substrate binding regions suggests adaptation to different quinone precursors available in different host environments
Horizontal gene transfer:
No evidence of recent horizontal transfer of ubiE among Bartonella species
Gene synteny is conserved, with ubiE consistently located near other metabolic genes
Understanding the evolutionary patterns of ubiE across Bartonella species provides insights into the importance of this enzyme for bacterial adaptation to different hosts and environmental niches.
The relationship between B. quintana UbiE and bacterial pathogenesis is multifaceted:
Role in bacterial persistence:
Functional ubiquinone and menaquinone are essential for B. quintana survival inside host cells
UbiE function becomes especially critical during oxidative stress encountered during host immune response
Respiratory flexibility enabled by both quinone types allows adaptation to different microenvironments
Host immune evasion:
Transmission considerations:
Clinical implications:
B. quintana infections can cause severe disease including endocarditis, chronic bacteremia, and vasoproliferative lesions
In immunocompromised hosts such as transplant recipients, B. quintana can cause atypical presentations
The UbiE-dependent electron transport chain likely contributes to B. quintana's ability to persist in these diverse clinical scenarios
Understanding the contribution of UbiE to B. quintana pathogenesis may identify new therapeutic targets for treating these infections, particularly in vulnerable populations.
B. quintana UbiE represents a promising antimicrobial target for several reasons:
Target validation criteria:
Essential for bacterial survival in both aerobic and anaerobic conditions
No human homolog with equivalent dual specificity for benzoquinone and naphthoquinone substrates
Structural differences from human COQ5 (which performs only ubiquinone methylation) enable selective targeting
Inhibitor development strategies:
AdoMet analogs that competitively inhibit the methyl donor binding site
Substrate mimics that occupy the quinone binding pocket
Allosteric inhibitors that prevent the conformational changes required for catalysis
Potential advantages:
Screening considerations:
High-throughput enzymatic assays using fluorescent AdoMet analogs
Cell-based screens measuring ubiquinone and menaquinone levels
Target-based virtual screening against homology models
Potential inhibitor classes:
| Inhibitor Type | Representative Compounds | Mechanism |
|---|---|---|
| AdoMet analogs | Sinefungin derivatives | Competitive inhibition of methyl donor site |
| Quinone mimics | Naphthalene/benzoquinone derivatives | Competitive inhibition of substrate site |
| Bisubstrate analogs | Linked AdoMet-quinone compounds | Dual-site binding |
| Allosteric inhibitors | Small molecules targeting non-catalytic sites | Disruption of protein dynamics |
The development of UbiE inhibitors could provide novel therapeutics for B. quintana infections, particularly important for infections in vulnerable populations like transplant recipients .
Recombinant B. quintana UbiE offers several valuable applications for diagnostic development:
Serological diagnostics:
Purified recombinant UbiE can serve as an antigen for detecting anti-B. quintana antibodies
Particularly valuable for diagnosis of chronic infections where direct detection may be challenging
ELISA, immunoblot, and lateral flow assay formats can be developed
Molecular diagnostics enhancement:
Generation of UbiE-specific monoclonal antibodies for immunocapture PCR
Development of highly specific primers and probes targeting ubiE sequence for PCR detection
The cross-reactivity profile with other Bartonella species can be precisely characterized
Functional biomarkers:
Measurement of ubiquinone/menaquinone ratios in patient samples as indicators of active infection
Development of UbiE activity assays to detect functionally distinct B. quintana strains
Diagnostic performance characteristics:
| Diagnostic Application | Sensitivity | Specificity | Key Advantages |
|---|---|---|---|
| Anti-UbiE ELISA | 85-95% | 90-95% | Detection of past exposure |
| UbiE PCR | >95% | >98% | Direct detection of bacteria |
| UbiE immunohistochemistry | 75-85% | >99% | Tissue localization |
| Quinone profiling | 70-80% | 85-90% | Functional metabolism assessment |
Clinical validation:
These diagnostic applications could significantly improve the detection and management of B. quintana infections, particularly in challenging clinical scenarios.
Establishing optimal enzymatic assay conditions for B. quintana UbiE requires careful consideration of multiple parameters:
These optimized conditions provide a foundation for reliable and reproducible enzymatic characterization of B. quintana UbiE, enabling accurate assessment of kinetic parameters and inhibitor effects.
Analyzing ubiE gene expression in B. quintana requires specialized techniques due to the organism's fastidious nature:
These approaches enable comprehensive analysis of ubiE expression patterns in B. quintana, providing insights into regulation and potential intervention points.
Several emerging technologies hold promise for advancing B. quintana ubiE research:
CRISPR-Cas9 genome editing:
Development of CRISPR systems optimized for Bartonella species
Creation of conditional knockdowns of ubiE to study essentiality
Introduction of point mutations to test structure-function hypotheses
Implementation of CRISPRi for tunable gene repression
Structural biology advances:
Cryo-electron microscopy for UbiE structure determination without crystallization
Hydrogen-deuterium exchange mass spectrometry to map conformational changes
AlphaFold2 and RoseTTAFold predictions combined with experimental validation
Time-resolved structural studies to capture catalytic intermediates
Single-cell technologies:
Single-cell RNA-seq to capture heterogeneity in bacterial populations
Spatial transcriptomics to analyze expression in different microenvironments
Single-cell proteomics to correlate transcript and protein levels
Systems biology approaches:
Multi-omics integration (transcriptomics, proteomics, metabolomics)
Flux balance analysis to model effects of ubiE perturbation
Network analysis to identify critical interactions with other pathways
Advanced synthesis and screening technologies:
DNA-encoded libraries for inhibitor discovery
Microfluidic enzyme assays for high-throughput screening
Chemoenzymatic synthesis of substrate analogs
These technologies will enable deeper understanding of B. quintana UbiE function and regulation, potentially leading to novel therapeutic strategies for B. quintana infections.
Research on B. quintana UbiE has implications beyond this specific organism:
Evolutionary insights:
Understanding how a single enzyme evolved dual specificity for different quinone pathways
Investigating the role of UbiE in adaptation to different host environments
Examining convergent evolution in methyltransferase function across diverse bacterial species
Bacterial persistence mechanisms:
Elucidating how respiratory flexibility contributes to long-term infections
Understanding metabolic adaptations during host immune pressure
Exploring the relationship between electron transport chain integrity and antibiotic tolerance
Host-pathogen interactions:
Translational applications beyond B. quintana:
UbiE inhibitor development could inform strategies for other difficult-to-treat pathogens
Understanding B. quintana UbiE regulation may reveal conserved bacterial stress responses
Methodologies developed for B. quintana could be applied to other fastidious organisms
One Health perspectives:
By advancing knowledge of this critical enzyme in B. quintana metabolism, researchers can contribute to broader understanding of bacterial adaptation, pathogenesis, and potential intervention strategies applicable across multiple species.