Essential for normal cell division and septation maintenance.
KEGG: dvu:DVU1664
STRING: 882.DVU1664
Desulfovibrio vulgaris has seen enormous progress in genetic manipulation techniques in recent years. For recombinant protein expression, researchers can utilize several systems, with the markerless genetic exchange system being particularly valuable. This system employs the uracil phosphoribosyltransferase gene (upp) as a counterselectable marker combined with 5-fluorouracil (5-FU) resistance for selection . The advantage of this approach is that it allows for multiple sequential genetic modifications without accumulating antibiotic resistance markers, which is crucial when engineering strains for recombinant protein expression .
For optimal expression of recombinant proteins like EngB, researchers should consider using the Δupp strain JW710, which has been developed specifically to facilitate genetic manipulation in D. vulgaris . This strain allows for a two-step integration and excision strategy that enables precise genetic modifications without polar effects that could interfere with protein expression.
The JW7035 strain, which contains deletions in both the upp gene and the hsdR gene (encoding a type I restriction endonuclease), demonstrates 100-1,000 times greater transformation efficiency compared to wild-type when introducing stable plasmids via electroporation . This makes it an excellent choice for recombinant EngB expression studies.
Transformation efficiency comparison for different D. vulgaris strains:
| D. vulgaris strain | Transformants/μg of plasmid DNA (pSC27) | Transformants/μg of plasmid DNA (pMO719) |
|---|---|---|
| Wild type | 2.1 × 10⁰ | 4.4 × 10⁰ |
| JW801 | 2.3 × 10² | 6.3 × 10³ |
| JW710 (Δupp) | 3.1 × 10⁻¹ | 3.2 × 10² |
| JW7035 (Δupp ΔhsdR) | 2.4 × 10³ | 2.8 × 10³ |
This data indicates that JW7035 is the optimal strain for introducing recombinant EngB constructs .
To determine the cellular localization of recombinant EngB in D. vulgaris, you can employ a GFP fusion approach similar to that used for studying CbiKᴾ localization . By fusing GFP to the C-terminus of EngB, you can visualize its localization using fluorescence microscopy.
The methodology involves:
Creating a genetic construct with the engB gene fused to gfp
Introducing this construct into an appropriate D. vulgaris strain (preferably JW7035 for higher transformation efficiency)
Confirming expression using Western blotting
Visualizing localization using fluorescence microscopy
Conducting subcellular fractionation to confirm microscopy results
This approach successfully confirmed the periplasmic localization of CbiKᴾ in previous studies and would be applicable to determining EngB localization .
The optimal protocol for constructing markerless deletion mutants to study EngB function in D. vulgaris involves a two-step integration and excision strategy using the upp/5-FU counterselection system. This methodology allows for the creation of clean, in-frame deletions without antibiotic markers that could interfere with subsequent genetic manipulations .
The detailed protocol includes:
Construct a suicide plasmid vector containing:
Approximately 1kb of DNA sequence upstream of engB
Approximately 1kb of DNA sequence downstream of engB
The wild-type upp gene expressed constitutively from a suitable promoter (e.g., the aph(3')-II promoter)
Transform the JW710 (Δupp) strain with this suicide plasmid vector
Select for integration of the plasmid (first recombination event) by screening for 5-FU sensitivity
Verify integration by PCR analysis
Select for excision of the plasmid (second recombination event) by plating on medium containing 5-FU
Screen 5-FU resistant colonies by PCR to identify those with the desired deletion
Confirm the deletion by sequencing
This approach has been successfully used to generate deletion strains in D. vulgaris with mutation frequencies of approximately 50% .
For purifying recombinant EngB from D. vulgaris for structural studies, a combination of approaches based on established protocols for similar proteins can be used:
Expression optimization:
Cell lysis and initial purification:
Use anaerobic techniques throughout purification due to D. vulgaris being an obligate anaerobe
Perform cell disruption by sonication or French press under anaerobic conditions
Clarify lysate by centrifugation at 15,000 × g for 30 minutes
Chromatography sequence:
Immobilized metal affinity chromatography (IMAC) as the initial capture step
Ion exchange chromatography for intermediate purification
Size exclusion chromatography as a final polishing step
Quality assessment:
SDS-PAGE to assess purity
Dynamic light scattering to assess homogeneity
Activity assays to confirm GTPase activity
Structure determination considerations:
Test protein stability in various buffers before crystallization attempts
Consider protein-GTP complexes for crystallization
Assess oligomerization state as it may affect crystal formation
This approach is based on successful purification methods used for other D. vulgaris proteins like CbiKᴾ, which was purified for structure-function relationship studies .
Site-directed mutagenesis for studying structure-function relationships in EngB can be optimized using a systematic approach similar to that employed for CbiKᴾ characterization . The methodology should focus on identifying key functional residues and determining their specific roles.
Optimized methodology includes:
In silico analysis to identify target residues:
Sequence alignment with homologous GTP-binding proteins to identify conserved residues
Structural prediction to identify residues in the GTP-binding pocket
Identification of potential metal coordination sites
Primer design considerations:
Use primers with a minimum of 20 bp complementary sequence flanking the mutation site
Ensure GC content of 40-60% for stable annealing
Verify primer specificity using in silico PCR tools
Mutagenesis protocol optimization:
Use high-fidelity polymerase to minimize unintended mutations
Optimize extension time based on template size
Consider using methylation-dependent selection to eliminate template DNA
Functional characterization of mutants:
GTPase activity assays to measure kinetic parameters (Km, Vmax)
Binding studies to assess GTP interaction (ITC, fluorescence-based assays)
Oligomerization analysis using size exclusion chromatography
Structural studies (if possible) to confirm the effects of mutations
This systematic approach has been successfully applied to D. vulgaris proteins, identifying key residues such as His154 and His216 in CbiKᴾ that are essential for its metal-chelation activity .
The deletion of the hsdR gene, encoding the type I restriction endonuclease in D. vulgaris, significantly impacts recombinant protein expression by enhancing transformation efficiency. Research has demonstrated that the JW7035 strain (Δupp ΔhsdR) exhibits 100-1,000 times greater transformation efficiency compared to wild-type D. vulgaris .
For recombinant EngB expression, this has several important implications:
Higher plasmid uptake efficiency leads to increased transformation success rates, reducing experimental variability and enhancing reproducibility .
The increased number of transformants allows for more efficient screening of expression constructs, facilitating optimization of expression conditions.
The absence of the restriction-modification system reduces the degradation of introduced DNA, allowing for more stable maintenance of expression vectors.
Transformation data indicates that with the JW7035 strain, researchers can expect approximately 2.4 × 10³ transformants/μg of plasmid DNA when using pSC27-based vectors, making it significantly more efficient than working with wild-type strains .
This enhancement in transformation efficiency is particularly valuable when working with challenging proteins like EngB, where expression optimization may require multiple rounds of construct modification and testing.
Resolving conflicting data regarding EngB oligomerization states requires a multi-technique approach that can provide complementary information. Based on methodologies used for studying other D. vulgaris proteins, the following strategies are recommended:
Size exclusion chromatography with multi-angle light scattering (SEC-MALS):
Provides direct measurement of absolute molecular weight independent of shape
Can detect multiple oligomeric species and their proportion in solution
Should be performed under various buffer conditions and protein concentrations
Analytical ultracentrifugation (AUC):
Sedimentation velocity experiments can resolve different oligomeric states
Provides information about shape and homogeneity
Less affected by protein-column interactions than SEC
Native mass spectrometry:
Can precisely determine oligomeric states and their distribution
Allows detection of ligand binding (e.g., GTP) that might influence oligomerization
Provides information about complex stability
Cross-linking mass spectrometry:
Identifies interaction interfaces between subunits
Can "freeze" transient interactions for analysis
Provides structural information about the oligomers
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Maps regions involved in protein-protein interactions
Can reveal conformational changes associated with oligomerization
Particularly useful when combined with mutagenesis studies
This comprehensive approach has been valuable in characterizing the tetrameric form of CbiKᴾ in D. vulgaris, identifying specific residues like Arg54 and Glu76 that stabilize the oligomeric structure through hydrogen bonding between subunits .
Identifying potential cofactors or binding partners of EngB in D. vulgaris requires a systematic approach combining in vivo and in vitro techniques. Based on successful methods used for studying D. vulgaris proteins, the following strategy is recommended:
Affinity purification coupled with mass spectrometry (AP-MS):
Express tagged EngB in D. vulgaris JW7035 strain
Perform gentle lysis under anaerobic conditions to preserve protein-protein interactions
Purify using an appropriate affinity tag (His-tag or FLAG-tag)
Identify co-purifying proteins by mass spectrometry
Validate interactions using reciprocal pull-downs
Bacterial two-hybrid screening:
Construct a D. vulgaris genomic library in a bacterial two-hybrid vector
Screen against EngB as bait
Validate positive interactions through secondary screening
Protein co-evolution analysis:
Perform bioinformatic analysis to identify proteins with similar phylogenetic profiles
Focus on proteins whose genes are consistently co-located with engB across bacterial genomes
Validate predicted interactions experimentally
Metabolite screening:
Use differential scanning fluorimetry to identify small molecules that stabilize EngB
Focus on GTP analogs and potential metal cofactors
Validate binding using isothermal titration calorimetry (ITC)
Crosslinking studies:
Perform in vivo crosslinking using formaldehyde or other crosslinkers
Isolate EngB complexes and identify crosslinked partners by mass spectrometry
This approach is inspired by successful studies of D. vulgaris proteins like CbiKᴾ, which identified unexpected cofactors such as haem binding through His96 and His103 residues that were absent in homologous proteins .
Low transformation efficiency is a significant challenge when working with D. vulgaris, but several strategies can help overcome this limitation:
Use optimized host strains:
Optimize electroporation conditions:
Use freshly prepared competent cells harvested in early to mid-log phase
Ensure complete removal of salts during washing steps
Optimize field strength, pulse duration, and temperature
Use pre-warmed recovery media immediately after electroporation
Plasmid considerations:
Use plasmids derived from the native D. vulgaris cryptic plasmid pBG1 for stable maintenance
Minimize plasmid size to improve transformation efficiency
Ensure plasmids are free of DNA methylation patterns recognized by D. vulgaris restriction systems
Protocol modifications:
Include a heat-shock step (42°C for 10 minutes) before electroporation
Add glycine betaine or DMSO as osmoprotectants during electroporation
Extend recovery time under anaerobic conditions
Comparative transformation efficiency data shows that with these optimizations, researchers can improve transformation rates from 2-5 transformants/μg plasmid DNA (wild-type) to over 2,000 transformants/μg (JW7035) .
Maintaining anaerobic conditions throughout the purification of recombinant EngB from D. vulgaris is crucial for preserving protein structure and activity. A comprehensive strategy includes:
Specialized equipment setup:
Use an anaerobic chamber or glove box maintained at <1 ppm O₂ for all purification steps
Equip the chamber with necessary chromatography systems and fraction collectors
Install a refrigeration unit within the chamber for temperature-sensitive steps
Use oxygen-scavenging catalysts and palladium catalysts to remove trace oxygen
Buffer preparation:
Degas all buffers by vacuum followed by sparging with nitrogen or argon
Include oxygen scavengers such as dithiothreitol (DTT) or β-mercaptoethanol
Consider adding sodium dithionite (0.5-2 mM) as a strong reducing agent
Pre-equilibrate buffers in the anaerobic chamber for at least 24 hours before use
Cell disruption considerations:
Harvest cells anaerobically and transfer to the chamber without exposure to oxygen
Use pressure-based lysis methods rather than sonication when possible
Maintain low temperature during lysis to prevent protein denaturation
Chromatography adaptations:
Use degassed mobile phases for all chromatography steps
Maintain a positive pressure of inert gas above buffer reservoirs
Keep flow rates moderate to minimize introduction of trace oxygen
Collect fractions directly into tubes containing additional oxygen scavengers
Activity preservation:
Perform activity assays within the anaerobic environment
Include GTP and appropriate metal ions in storage buffers to stabilize EngB
Consider flash-freezing aliquots in liquid nitrogen within the chamber for long-term storage
This comprehensive approach is essential for maintaining the native structure and function of oxygen-sensitive proteins from anaerobic organisms like D. vulgaris .
Differentiating between properly folded and misfolded recombinant EngB requires a multi-technique analytical approach that examines different aspects of protein structure and function:
Functional assays:
GTPase activity measurements using colorimetric or fluorescence-based assays
GTP binding assays using fluorescent GTP analogs
Comparison of kinetic parameters (Km, kcat) with those of native or homologous proteins
Spectroscopic techniques:
Circular dichroism (CD) spectroscopy to assess secondary structure content
Fluorescence spectroscopy to monitor tertiary structure through intrinsic tryptophan fluorescence
Fourier-transform infrared spectroscopy (FTIR) to detect aggregation and non-native β-sheet formation
Hydrodynamic methods:
Size-exclusion chromatography to detect aggregation or aberrant oligomerization
Dynamic light scattering to assess size distribution and polydispersity
Analytical ultracentrifugation to determine shape parameters and oligomeric state
Thermal stability analysis:
Differential scanning calorimetry (DSC) to measure conformational stability
Differential scanning fluorimetry (DSF) using environment-sensitive dyes
Temperature-dependent activity assays to correlate structure with function
Protease susceptibility:
Limited proteolysis followed by mass spectrometry to identify exposed, potentially misfolded regions
Comparison of digestion patterns between purified protein and native controls
This approach has been valuable in characterizing properly folded D. vulgaris proteins, as demonstrated in structure-function studies of CbiKᴾ, where both functional assays and structural analyses were used to confirm proper protein folding after mutagenesis .
Cryo-electron microscopy (cryo-EM) offers significant advantages for studying EngB structure and interactions, particularly for capturing dynamic states. Optimizing cryo-EM for this specific application requires addressing several key considerations:
Sample preparation optimization:
Protein concentration: Test concentration ranges (typically 0.5-5 mg/ml) to identify optimal particle distribution
Buffer composition: Screen different buffers to minimize background and maximize contrast
Grid treatment: Test different grid types (Quantifoil, C-flat) and surface treatments (glow discharge parameters, graphene coating)
Vitrification conditions: Optimize blotting time and temperature to achieve ideal ice thickness
Data collection strategy:
Use energy filters to improve signal-to-noise ratio for a relatively small protein like EngB
Implement beam-tilt data collection to increase throughput and improve reconstruction quality
Consider collecting data in super-resolution mode with subsequent binning
Use beam-image shift strategy for faster data collection while maintaining quality
EngB-specific considerations:
Capture different nucleotide-bound states (GDP vs. GTP vs. apo)
Study EngB in complex with interaction partners identified through pull-down experiments
Use GTPase-deficient mutants to trap transition states
Consider using nanodiscs to study membrane-associated forms if relevant
Processing workflow optimization:
Implement 2D classification strategies suitable for smaller proteins
Use 3D classification to separate different conformational states
Apply focused refinement on regions of interest
Consider Bayesian polishing and CTF refinement to maximize resolution
This approach builds on techniques used for structural studies of other D. vulgaris proteins, adapting them to the specific challenges of EngB as a probable GTP-binding protein with potential dynamic conformational states .
Comparative genomics can provide valuable insights into the evolution and function of EngB across Desulfovibrio species, revealing conservation patterns, potential functional adaptations, and evolutionary relationships:
Conservation analysis:
Multiple sequence alignment of EngB proteins across Desulfovibrio species and other bacterial genera
Identification of universally conserved residues that likely play critical roles in GTP binding and hydrolysis
Mapping conservation patterns onto structural models to identify functional surfaces
Analysis of selection pressure (dN/dS ratios) to identify residues under positive or purifying selection
Genomic context analysis:
Examination of gene neighborhoods around engB across different Desulfovibrio species
Identification of consistently co-localized genes that may be functionally related
Comparison with other sulfate-reducing bacteria to identify shared genomic architectures
Investigation of potential operon structures and co-regulation patterns
Phylogenetic profiling:
Construction of phylogenetic trees based on EngB sequences
Correlation with species phylogeny to identify potential horizontal gene transfer events
Identification of lineage-specific adaptations in EngB structure and function
Correlation of EngB variants with ecological niches of different Desulfovibrio species
Structural variation analysis:
Prediction of structural differences in EngB proteins across Desulfovibrio species
Identification of insertions/deletions that may confer specialized functions
Analysis of surface properties and potential interaction interfaces
Correlation of structural variations with biochemical characteristics of different species
This comprehensive comparative genomics approach draws inspiration from studies of other D. vulgaris proteins, such as CbiKᴾ, which revealed unique evolutionary adaptations like the acquisition of haem-binding capabilities through specific histidine residues (His96 and His103) that are absent in homologous proteins from other bacteria .
Integrating EngB function into the broader metabolic network of D. vulgaris requires sophisticated system-wide approaches that combine multiple omics technologies with computational modeling:
Multi-omics data integration:
Transcriptomics: Analyze co-expression patterns between engB and other genes under various conditions
Proteomics: Identify changes in the proteome when engB is deleted or overexpressed
Metabolomics: Measure metabolic shifts associated with altered EngB function
Interactomics: Map protein-protein interactions involving EngB using techniques like AP-MS
Network analysis approaches:
Construct protein-protein interaction networks centered on EngB
Develop gene regulatory networks to identify factors controlling engB expression
Create metabolic flux models incorporating EngB-dependent processes
Identify network motifs and modules associated with EngB function
Perturbation studies:
Generate conditional engB mutants using the markerless deletion system
Perform time-resolved multi-omics analyses after EngB depletion
Conduct synthetic lethality screens to identify genes functionally related to engB
Implement CRISPR interference for targeted down-regulation of engB and potential partners
Computational model development:
Integrate experimental data into genome-scale metabolic models of D. vulgaris
Perform flux balance analysis with and without functional EngB
Develop kinetic models of GTP-dependent processes involving EngB
Use machine learning approaches to predict condition-specific roles of EngB
This systems biology approach builds on the genetic manipulation techniques available for D. vulgaris, particularly the markerless deletion system that allows for the creation of clean genetic backgrounds suitable for system-wide studies . The approach is particularly valuable for understanding proteins like EngB that may have multiple cellular roles or participate in complex regulatory networks.
Optimizing expression of soluble, active recombinant EngB in D. vulgaris requires a systematic approach addressing multiple aspects of protein production:
Strain selection and optimization:
Expression construct design:
Test different promoter systems (constitutive vs. inducible)
Optimize the ribosome binding site sequence for efficient translation
Consider codon optimization based on D. vulgaris codon usage
Evaluate N-terminal vs. C-terminal tags based on protein topology
Include a TEV protease cleavage site for tag removal
Expression condition optimization:
Test different growth temperatures (typically 30-37°C)
Evaluate various induction times and durations
Optimize cell density at induction (mid-log vs. late-log phase)
Supplement growth media with appropriate cofactors (GTP, metal ions)
Solubility enhancement strategies:
Co-express with molecular chaperones if folding issues are observed
Test fusion partners known to enhance solubility (SUMO, MBP, TrxA)
Evaluate the effect of osmolytes in the growth medium
Consider periplasmic targeting if appropriate
Purification strategy optimization:
Maintain anaerobic conditions throughout purification
Include stabilizing agents (GTP, metal ions) in all buffers
Optimize purification buffer composition (pH, salt concentration, additives)
Implement a multi-step purification strategy tailored to EngB properties
This systematic approach draws on the successful genetic manipulation techniques developed for D. vulgaris, particularly the advanced transformation systems that allow for efficient introduction of expression constructs .
Adapting in vivo imaging techniques to study EngB localization and dynamics in D. vulgaris requires specialized approaches that account for both the anaerobic nature of the organism and its specific biology:
Fluorescent protein optimization:
Test oxygen-independent fluorescent proteins (e.g., flavin-based fluorescent proteins)
Evaluate split-GFP systems where one fragment is fused to EngB and the other expressed separately
Consider photoconvertible fluorescent proteins for pulse-chase experiments
Validate that the GFP fusion approach successfully used for CbiKᴾ localization is also suitable for EngB
Microscopy setup adaptation:
Implement anaerobic chambers that interface with microscope stages
Use sealed, gas-impermeable imaging chambers with oxygen scavengers
Optimize illumination parameters to minimize phototoxicity
Consider light sheet microscopy for reduced photobleaching and photodamage
Dynamic imaging approaches:
Employ fluorescence recovery after photobleaching (FRAP) to measure protein mobility
Use single-particle tracking for studying EngB movement within cells
Implement fluorescence correlation spectroscopy (FCS) to measure diffusion rates
Apply stimulated emission depletion (STED) microscopy for super-resolution imaging
Multi-color imaging strategies:
Co-express EngB fusion with markers for different cellular compartments
Study co-localization with potential interaction partners
Use fluorescence resonance energy transfer (FRET) to detect protein-protein interactions
Implement fluorescent timers to track protein age and turnover
Data analysis considerations:
Develop automated image analysis pipelines specific for D. vulgaris cell morphology
Implement deconvolution algorithms optimized for anaerobic imaging conditions
Use machine learning approaches for feature detection and classification
Apply quantitative analysis methods to extract kinetic parameters
This approach builds on the successful C-terminal GFP fusion strategy that confirmed the periplasmic localization of CbiKᴾ in D. vulgaris, adapting it for the specific requirements of studying EngB dynamics .
Measuring GTPase activity of recombinant EngB under strictly anaerobic conditions requires specialized assays that can function in the absence of oxygen. The following approaches are recommended:
Colorimetric phosphate detection methods:
Malachite green assay:
Can be performed anaerobically in a glove box
Measures free phosphate released during GTP hydrolysis
Sample protocol:
a) Incubate purified EngB (0.1-5 μM) with GTP (0.1-1 mM) in reaction buffer
b) At timed intervals, remove aliquots and add to malachite green solution
c) Measure absorbance at 630 nm after color development
Include standard curves prepared under identical conditions
PiColorLock Gold assay:
Higher sensitivity than malachite green
Lower interference from reducing agents commonly used in anaerobic buffers
Can be adapted for microplate format for higher throughput
HPLC-based nucleotide analysis:
Separate and quantify GTP and GDP using ion-pair reversed-phase HPLC
Sample preparation can be performed anaerobically with analysis under aerobic conditions
Allows direct measurement of substrate consumption and product formation
Can detect other potential nucleotide intermediates
Coupled enzyme assays:
Pyruvate kinase/lactate dehydrogenase coupled system:
Links GTP hydrolysis to NADH oxidation (measurable at 340 nm)
All enzymes must be oxygen-stable or used within an anaerobic chamber
Requires confirmation that coupling enzymes aren't affected by anaerobic conditions
Fluorescence-based methods:
BODIPY-GTP assays:
Measures changes in fluorescence upon GTP hydrolysis
Can be performed in sealed, oxygen-impermeable cuvettes
Allows continuous, real-time monitoring of activity
Radioactive assays:
[γ-³²P]GTP hydrolysis:
Extremely sensitive detection of GTPase activity
Can be performed under strictly anaerobic conditions
Separation of radioactive Pi from GTP by thin-layer chromatography
Requires appropriate radioactive material handling facilities
These methods should be calibrated using known GTPase proteins and appropriate controls to ensure reliability under anaerobic conditions. The approach is informed by successful enzymatic assays developed for other D. vulgaris proteins, adapted for the specific requirements of measuring GTPase activity .
Adapting CRISPR-Cas9 genome editing for precise manipulation of engB in D. vulgaris would represent a significant advancement over current genetic manipulation techniques. The following approach outlines how this could be accomplished:
Development of a D. vulgaris-optimized CRISPR-Cas9 system:
Design considerations for targeting engB:
Analyze the engB locus for optimal protospacer adjacent motif (PAM) sites
Design gRNAs with minimal off-target effects across the D. vulgaris genome
Create repair templates that include desired modifications (point mutations, tags, etc.)
Consider using nickase variants for higher specificity
Integration with existing genetic systems:
Validation and optimization:
Develop efficient screening methods to identify successful edits
Optimize homology-directed repair efficiency through manipulation of DNA repair pathways
Establish protocols for multiplexed editing of engB and related genes
Create a library of validated gRNAs for different regions of engB
Advanced applications:
Develop CRISPRi systems for tunable repression of engB expression
Create CRISPRa systems for enhanced expression when needed
Establish base editing capabilities for precise single nucleotide modifications
Develop protocols for large-scale functional genomics studies
This approach would significantly advance beyond the current markerless deletion system by offering more precise and versatile genetic manipulation capabilities for studying engB function in D. vulgaris .
Detailed structural and functional characterization of EngB from D. vulgaris could lead to several innovative biotechnological applications:
Novel antimicrobial development:
If EngB proves essential for D. vulgaris survival, structural data could guide the design of selective inhibitors
Comparative analysis with homologs from pathogenic bacteria could identify conserved targetable features
Structure-based virtual screening could identify compounds that interfere with GTP binding
Rational design of peptide inhibitors targeting unique structural features of bacterial EngB proteins
Protein engineering applications:
Development of GTP-responsive molecular switches based on EngB conformational changes
Creation of biosensors for detecting GTP or related nucleotides in complex biological samples
Engineering EngB variants with altered nucleotide specificity for specialized applications
Design of EngB-based scaffold proteins for organizing multi-enzyme complexes
Biocatalysis innovations:
Exploration of potential promiscuous enzymatic activities that could be enhanced through directed evolution
Development of EngB variants capable of catalyzing novel chemical transformations
Creation of chimeric enzymes combining EngB domains with other catalytic modules
Engineering oxygen-tolerant variants for broader biotechnological applications
Bioremediation applications:
If EngB plays a role in stress responses, engineered variants could enhance D. vulgaris survival in contaminated environments
Development of biosensors for monitoring environmental conditions based on EngB activity
Creation of immobilized EngB systems for specific biotransformations relevant to environmental cleanup
Engineering D. vulgaris strains with enhanced EngB function for improved metal precipitation or reduction
Structural biology tools:
Development of EngB as a crystallization chaperone for difficult-to-crystallize proteins
Creation of EngB-based affinity tags for protein purification under anaerobic conditions
Design of EngB fusion proteins for enhancing solubility of recalcitrant proteins
Development of engineered EngB domains as molecular probes for studying GTP-dependent processes
These applications would build upon the detailed structural and functional insights gained from studies similar to those conducted on other D. vulgaris proteins like CbiKᴾ, where unexpected functions (haem binding) were discovered through rigorous structural and biochemical analysis .
Synthetic biology approaches offer exciting possibilities for engineering novel functions into EngB from D. vulgaris, potentially creating proteins with unique and valuable properties:
Domain fusion engineering:
Create chimeric proteins by fusing EngB with domains from other proteins:
EngB-fluorescent protein fusions for in vivo biosensing applications
EngB-enzyme fusions for GTP-dependent catalysis regulation
EngB-binding domain fusions for controlled protein recruitment
Optimize linker regions between domains for optimal function
Test various domain orientations to minimize steric hindrance
Directed evolution strategies:
Develop high-throughput screening systems compatible with anaerobic conditions
Apply error-prone PCR to generate EngB variant libraries
Use PACE (phage-assisted continuous evolution) adapted for D. vulgaris
Implement targeted mutagenesis of GTP-binding regions to alter nucleotide specificity
Select for variants with novel properties such as:
Altered nucleotide specificity (ATP, UTP instead of GTP)
Enhanced stability under various conditions
New catalytic activities
Computational design approaches:
Use Rosetta-based computational tools to redesign the active site
Apply machine learning algorithms to predict beneficial mutations
Design de novo binding sites for novel ligands
Engineer allosteric regulation mechanisms into EngB structure
Genetic circuit integration:
Incorporate engineered EngB variants into synthetic genetic circuits
Design GTP-responsive gene expression systems using EngB as a sensor
Create oscillatory circuits based on EngB activity
Develop complex logical operations using multiple engineered EngB variants
Non-canonical amino acid incorporation:
Introduce reactive handles for bioorthogonal chemistry
Incorporate photocrosslinkable amino acids for controlled protein interactions
Add fluorescent amino acids for direct activity monitoring
Introduce metal-binding residues for novel catalytic activities
This synthetic biology approach would build upon the genetic manipulation tools available for D. vulgaris, particularly the markerless deletion system that allows for clean genetic backgrounds suitable for synthetic biology applications . The approach is also informed by successful protein engineering studies like those conducted on CbiKᴾ, where specific residues (His96, His103) were identified as critical for novel functions .