The protein is synthesized using recombinant DNA technology in two primary systems:
Host: Insect cells for eukaryotic post-translational modifications .
Storage: Liquid or lyophilized forms with a shelf life of 6–12 months at -80°C .
The crgA gene resides in a highly plastic genomic region of T. whipplei, characterized by recombination hotspots involving conserved repeats . These rearrangements may alter surface protein expression, potentially aiding immune evasion . Key genomic data:
Genome Feature | T. whipplei Twist Strain |
---|---|
Genome Size | 927,303 bp |
G+C Content | 46.3% |
Coding Density | 85.6% |
Protein-Coding Genes | 808 |
T. whipplei lacks biosynthetic pathways for several amino acids, necessitating host-dependent nutrient uptake . TWT_010’s role in membrane dynamics could facilitate this process, though direct evidence remains limited .
Antigen Production: Used to generate antibodies for detecting T. whipplei in clinical samples .
Structural Studies: The conserved WND-domain repeats are of interest for understanding bacterial genome plasticity .
Diagnostic Development: Potential use in serological assays, given its membrane localization .
KEGG: twh:TWT_010
STRING: 203267.TWT010
Tropheryma whipplei is the bacterial agent responsible for Whipple's disease, a chronic condition first described in 1907 by Nobel laureate George Whipple. This disease manifests as intestinal malabsorption leading to cachexia and can be fatal without appropriate antibiotic intervention. T. whipplei is particularly notable as a human pathogen with a significantly reduced genome (<1 Mb), making it unique among the Actinobacteria phylum (high G+C Gram-positive bacteria). The bacterium resisted reproducible culture until 2000 when it was successfully grown in human fibroblasts. Physically, T. whipplei is a small bacterium (0.3 × 1.5 mm) that stains Gram-negative, possesses an atypical envelope, and features a thick cell wall. In culture, it exhibits distinctive giant rope-like structures similar to those seen in Mycobacterium tuberculosis .
TWT_010 is classified as a UPF0233 family membrane protein encoded within the 927,303-bp circular genome of T. whipplei Twist strain. The complete genome encodes 808 predicted protein-coding genes and 54 RNA genes, with an average G+C content of 46% (significantly lower than other high G+C content Gram-positive bacteria). The coding content of the entire genome is approximately 85.6%. TWT_010 belongs to the membrane protein complement that comprises part of this highly compact genome. The protein is designated by its locus identifier based on its position in the annotated genome sequence of the Twist strain .
TWT_010 belongs to the UPF0233 family of membrane proteins, which typically feature transmembrane domains that anchor the protein within the bacterial cell envelope. While the specific structure of TWT_010 has not been fully resolved, membrane proteins in T. whipplei generally contain hydrophobic amino acid sequences that form transmembrane helices. Based on comparative analysis with other bacterial membrane proteins, TWT_010 likely contains multiple transmembrane domains with intervening loops that may be exposed to either the cytoplasmic or extracellular environment. The protein may also contain specific functional domains related to its biological role, possibly including regions involved in protein-protein interactions or substrate binding .
The genomic context of TWT_010 may provide clues about its function through examination of nearby genes and their predicted functions. T. whipplei has numerous genes encoding various biological functions despite its reduced genome size compared to other bacteria with similarly small genomes (<1 Mb). The bacterium exhibits genomic inversions between different strains (such as between Twist and TW08/27), with the inversion boundaries located within paralogous genes of cell-surface protein families. This genomic plasticity may affect the expression of membrane proteins including TWT_010, potentially influencing the bacterium's surface protein complement. By analyzing genes adjacent to TWT_010 and their conservation across different T. whipplei strains, researchers may gain insights into potential operonic structures and functional relationships .
For recombinant production of T. whipplei TWT_010 membrane protein, several expression systems should be considered, each with specific advantages:
E. coli-based systems: The BL21(DE3) strain with pET vector systems provides high-yield expression, though membrane proteins often form inclusion bodies requiring refolding. Consider using C41(DE3) or C43(DE3) strains specifically engineered for membrane protein expression.
Cell-free expression systems: These bypass toxicity issues and allow direct incorporation into nanodiscs or liposomes for functional studies.
QTY code-modified expression: Applying the QTY code substitution strategy, where hydrophobic residues in transmembrane domains (leucine, isoleucine, valine) are replaced with hydrophilic residues (glutamine, threonine, tyrosine) while maintaining similar side chain size and structure. This approach renders membrane proteins water-soluble without detergents, potentially simplifying purification while maintaining structural integrity and ligand-binding capabilities .
For optimal results, expression trials should include different promoter strengths, induction temperatures (16-30°C), and inducer concentrations. Fusion tags (such as MBP, SUMO, or thioredoxin) can enhance solubility and facilitate detection and purification.
Purification of TWT_010 membrane protein presents significant challenges requiring specialized approaches. The following strategy is recommended for optimal results:
Membrane extraction: Solubilize membranes using mild detergents such as n-dodecyl-β-D-maltoside (DDM), lauryl maltose neopentyl glycol (LMNG), or digitonin to maintain protein structure.
Affinity chromatography: Utilize N-terminal or C-terminal affinity tags (His6, FLAG, or Strep-II) for initial purification, with optimized imidazole gradients to minimize non-specific binding.
Size exclusion chromatography: Remove aggregates and further purify monomeric protein.
QTY code application: If using QTY-modified TWT_010, conventional aqueous buffer purification can be employed without detergents, significantly simplifying the process .
Purification Step | Traditional Approach | QTY-Modified Approach |
---|---|---|
Extraction | Detergent solubilization (DDM, LMNG) | Aqueous buffer extraction |
Initial Capture | IMAC with detergent-containing buffers | Standard IMAC without detergents |
Intermediate Purification | Ion exchange with detergent | Standard ion exchange |
Polishing | Size exclusion with detergent | Standard size exclusion |
Final Storage | Detergent concentration above CMC | Standard aqueous buffer |
The QTY code approach may eliminate the need for detergents throughout the purification process, potentially improving yield and simplifying downstream applications .
Structural characterization of TWT_010 requires a multi-technique approach to overcome the challenges inherent in membrane protein analysis:
X-ray crystallography:
Screen various detergents and lipid additives to identify conditions promoting crystal formation
Consider lipidic cubic phase (LCP) crystallization, which mimics the membrane environment
Use fusion partners like BRIL or T4 lysozyme to enhance crystal contacts
Cryo-electron microscopy (cryo-EM):
Particularly valuable if TWT_010 forms multimeric complexes
Use nanodiscs or amphipols to maintain native-like membrane environment
Apply different grid preparation techniques (continuous carbon, holey carbon with thin ice)
NMR spectroscopy:
For QTY-modified TWT_010, solution NMR becomes feasible without micelle interference
Selective isotope labeling can facilitate assignment of resonances
Deuteration may improve spectral quality for this medium-sized protein
Computational approaches:
Homology modeling based on structurally characterized UPF0233 family proteins
Molecular dynamics simulations to predict stability in membrane environments
AlphaFold2 or RoseTTAFold predictions validated by limited experimental data
The QTY code modification strategy can significantly simplify structural studies by eliminating detergent micelles that often complicate data collection and analysis in traditional membrane protein structural biology .
Functional characterization of TWT_010 requires assays relevant to its predicted membrane-associated roles:
Lipid interaction assays:
Monolayer insertion measurements
Fluorescence-based binding assays with labeled lipids
Surface plasmon resonance with immobilized lipid bilayers
Protein-protein interaction studies:
Pull-down assays with potential partner proteins
Yeast two-hybrid with modified membrane segments
FRET-based interaction assays
Transport activity (if TWT_010 functions as a transporter):
Liposome reconstitution with fluorescent substrates
Patch-clamp electrophysiology if ion transport is suspected
Substrate uptake assays in whole cells overexpressing TWT_010
Cell-based functional assays:
Complementation of knockout mutants
Localization studies using fluorescent protein fusions
Impact on bacterial survival under various stresses
When using QTY-modified versions for functional studies, it's essential to validate that the substitutions don't significantly alter the protein's functional properties by comparing results with detergent-solubilized wild-type protein wherever possible .
T. whipplei exhibits significant genomic plasticity, with a notable large chromosomal inversion observed between the Twist and TW08/27 strains. This inversion's boundaries are located within paralogous genes belonging to a cell-surface protein family characterized by highly conserved nucleotide repeats. These repeats appear to trigger frequent genome rearrangements in T. whipplei, potentially resulting in differential expression of cell surface proteins. This mechanism may represent a novel strategy for evading host immune defenses .
For TWT_010 specifically, researchers should investigate:
Positional effects: Determine if TWT_010's genomic location is conserved across strains or affected by inversions, which could alter its expression through changes in promoter context or operon structure.
Transcriptional analysis: Compare TWT_010 mRNA levels across different T. whipplei strains using RT-qPCR and RNA-seq to identify strain-specific expression patterns.
Regulatory elements: Analyze the upstream regions of TWT_010 across strains to identify potential regulatory sequences that might be affected by genomic rearrangements.
Co-expression networks: Investigate whether genomic rearrangements alter co-expression patterns between TWT_010 and other genes, potentially indicating functional relationships.
Understanding how genomic plasticity affects TWT_010 expression could provide insights into both the protein's function and the bacterium's adaptive strategies.
Identifying TWT_010's interacting partners is crucial for understanding its function within T. whipplei. Several complementary approaches are recommended:
Co-immunoprecipitation with mass spectrometry:
Express tagged TWT_010 in T. whipplei or heterologous systems
Cross-link protein complexes in vivo before extraction
Identify pulled-down proteins via LC-MS/MS
Validate interactions with reciprocal pull-downs
Bacterial two-hybrid systems:
Construct fusion proteins with split adenylate cyclase domains
Screen against a genomic library of T. whipplei
Validate positive interactions with targeted assays
Proximity labeling approaches:
Fuse TWT_010 with BioID or APEX2 enzymes
Identify proteins in proximity through biotinylation
Analyze spatial relationships in the membrane context
Computational prediction:
Use protein-protein interaction algorithms
Identify conserved interaction motifs
Employ coevolution analysis across related species
Interaction Method | Advantages | Limitations | Best For |
---|---|---|---|
Co-IP/MS | Captures native complexes | Requires good antibodies | Stable interactions |
Bacterial Two-Hybrid | Scalable screening | High false positive rate | Binary interactions |
Proximity Labeling | Captures transient interactions | Spatial resolution limited | In vivo context |
Computational | Rapid, low cost | Requires validation | Generating hypotheses |
Combining these approaches provides a robust strategy for mapping TWT_010's interactome, offering insights into its cellular function.
Investigating TWT_010's potential role in pathogenesis requires approaches that connect molecular function with disease processes:
Gene knockout or knockdown studies:
Develop CRISPR-Cas or antisense RNA systems for T. whipplei
Assess impact on bacterial survival in macrophages
Evaluate changes in inflammatory responses
Infection models:
Establish cell culture infection systems with wild-type and TWT_010-deficient strains
Measure adhesion, invasion, and intracellular survival
Assess host cell responses through transcriptomics and cytokine profiling
Immunological studies:
Determine if TWT_010 is recognized by antibodies from Whipple's disease patients
Assess T cell responses to TWT_010 epitopes
Evaluate inflammatory potential of purified TWT_010
Structural mimicry analysis:
Investigate potential molecular mimicry between TWT_010 and host proteins
Identify regions that could interfere with host signaling pathways
Explore cross-reactivity with host epitopes
These approaches can help determine whether TWT_010 plays a direct role in Whipple's disease pathogenesis, potentially identifying new therapeutic targets or diagnostic markers.
Computational structure predictions for TWT_010 require experimental validation through multiple techniques:
Limited proteolysis:
Identify protease-resistant domains indicating structured regions
Compare experimental fragments with predicted domain boundaries
Use mass spectrometry to precisely map cleavage sites
Disulfide mapping:
Introduce cysteine pairs at predicted proximal residues
Assess disulfide formation under oxidizing conditions
Validate predicted 3D proximity relationships
Epitope mapping:
Generate antibodies against predicted exposed regions
Confirm accessibility through binding studies
Use mutational analysis to validate epitope locations
DEER spectroscopy:
Introduce spin labels at specific residues
Measure distances between labeled sites
Compare with distances predicted in computational models
Hydrogen-deuterium exchange mass spectrometry:
Identify regions with differential solvent exposure
Compare with predicted surface accessibility
Map protein dynamics to predicted structural elements
An integrated approach combining these methods provides robust validation of structural predictions, especially important given the challenges of obtaining high-resolution experimental structures of membrane proteins like TWT_010.
Expression of membrane proteins like TWT_010 frequently encounters challenges including toxicity, low yield, and improper folding. The following strategies can help overcome these barriers:
Controlling expression levels:
Use titratable promoter systems (tetracycline-inducible, rhamnose-inducible)
Optimize ribosome binding sites to modulate translation efficiency
Consider low-copy number plasmids to reduce metabolic burden
Expression strain engineering:
Utilize C41/C43(DE3) strains optimized for membrane protein expression
Consider Lemo21(DE3) with tunable T7 lysozyme levels
Select strains with enhanced oxidative folding capability for disulfide-containing domains
QTY code implementation:
Fusion partner screening:
Test multiple fusion partners (MBP, SUMO, Trx) at N- and C-termini
Optimize linker lengths between TWT_010 and fusion partners
Include precise protease cleavage sites for tag removal
Strategy | Implementation | Expected Outcome |
---|---|---|
Reduced expression rate | Lower temperature (16-20°C), weaker promoters | Improved folding, less aggregation |
Specialized host strains | C41/C43(DE3), SHuffle | Reduced toxicity, better folding |
QTY code modification | L→Q, I→Q, V→T substitutions | Water-soluble variant without detergents |
Fusion tags | N-terminal MBP, SUMO | Enhanced solubility, yield |
The QTY code approach represents a particularly innovative solution, as it can transform the inherently hydrophobic membrane protein into a water-soluble variant while preserving structural features and functional properties .
Maintaining stability of purified TWT_010 is critical for downstream functional and structural studies. Several approaches can be implemented:
Buffer optimization:
Screen pH ranges (typically 6.0-8.0) for optimal stability
Test various salt concentrations (100-500 mM) and types (NaCl, KCl)
Include stabilizing additives (glycerol 5-20%, sucrose, arginine)
Detergent selection (for native TWT_010):
Screen detergent panels (maltoside, glucoside, and neopentyl glycol families)
Consider detergent mixtures for enhanced stability
Optimize detergent concentration just above critical micelle concentration
Lipid supplementation:
Add specific phospholipids (POPC, POPE, cardiolipin) to mimic bacterial membrane
Use cholesterol hemisuccinate as a stabilizing agent
Consider nanodiscs or SMALPs for native-like lipid environment
QTY-modified approach:
Storage considerations:
Test protein stability at different temperatures (4°C, -20°C, -80°C)
Evaluate flash-freezing in liquid nitrogen vs. slow freezing
Consider lyophilization for long-term storage of QTY-modified variants
Systematic stability screening using techniques like differential scanning fluorimetry (DSF) or thermal shift assays can efficiently identify optimal stabilization conditions.
Protein aggregation represents a major challenge in membrane protein purification, including TWT_010. Several targeted strategies can mitigate this issue:
Prevention during cell lysis:
Maintain cold temperatures (4°C) throughout processing
Include protease inhibitors to prevent degradation-induced aggregation
Add reducing agents (DTT, TCEP) to prevent disulfide-mediated aggregation
Solubilization optimization:
Screen detergent:protein ratios to ensure complete solubilization
Allow sufficient solubilization time (4-16 hours) with gentle agitation
Remove insoluble material via ultracentrifugation (100,000×g)
Chromatography considerations:
Use moderate flow rates to reduce shear stress
Maintain detergent above CMC in all purification buffers
Consider on-column detergent exchange during affinity steps
QTY code advantages:
Analytical quality control:
Monitor aggregation state via dynamic light scattering
Use size exclusion chromatography with multi-angle light scattering (SEC-MALS)
Implement regular thermal stability checks during purification
Implementing these strategies systematically can significantly reduce aggregation issues, improving yield and homogeneity of purified TWT_010 for downstream applications.
Robust control experiments are essential for reliable interpretation of TWT_010 functional studies:
Negative controls:
Empty vector/untransfected cells for expression studies
Heat-denatured TWT_010 for activity assays
Unrelated membrane protein of similar size and topology
Liposomes without reconstituted protein
Positive controls:
Well-characterized membrane protein from same family (if available)
Known functional domains expressed independently
Chemical controls for assay validation
QTY-specific controls:
Validation approaches:
Multiple independent protein preparations
Different expression systems for cross-validation
Concentration-dependent activity measurements
Multiple independent assay methodologies
Control Type | Purpose | Implementation |
---|---|---|
Negative | Establish baseline, detect artifacts | Empty vector, denatured protein |
Positive | Validate assay function | Known functional protein, chemical controls |
QTY-specific | Assess impact of modifications | Wild-type vs. QTY variant comparison |
Technical | Ensure reproducibility | Multiple preparations, concentration series |
Through systematic implementation of these controls, researchers can confidently attribute observed activities to TWT_010 and distinguish between native function and artifacts introduced by expression systems or purification methods.
Structural characterization of TWT_010 could significantly advance understanding of T. whipplei biology and pathogenesis through several key contributions:
Functional insights:
Identification of potential binding pockets or catalytic sites
Structural homology to proteins of known function
Mapping of conserved residues to functional domains
Drug development potential:
Identification of druggable pockets specific to TWT_010
Structure-based design of inhibitors or modulators
Virtual screening campaigns against the solved structure
Host-pathogen interaction understanding:
Mapping epitopes recognized by host immune system
Identifying regions involved in host cell attachment
Understanding mechanisms of immune evasion
Membrane protein biology advances:
Contributions to UPF0233 family structural knowledge
Understanding membrane protein adaptation in reduced genomes
Novel structural motifs unique to this specialized pathogen
The QTY code approach could significantly facilitate structural studies by enabling solution-phase techniques without the complications of detergent micelles, potentially accelerating progress in this challenging area of research .
Advanced genomic approaches offer powerful tools for understanding TWT_010 function:
Comparative genomics:
Analysis across T. whipplei strains for conservation patterns
Examination of UPF0233 family proteins across bacterial species
Identification of co-evolving gene clusters suggesting functional relationships
Transcriptomic profiling:
RNA-seq under various growth conditions and stresses
Co-expression network analysis to identify functional modules
Differential expression during infection of host cells
Genetic manipulation strategies:
CRISPR-Cas9 approaches for precise genome editing
Transposon mutagenesis libraries with TWT_010 knockouts
Complementation studies with mutant variants
Metatranscriptomics of clinical samples:
Expression analysis directly from Whipple's disease specimens
Correlation of TWT_010 expression with disease severity
Identification of in vivo regulatory mechanisms
These genomic approaches can provide contextual understanding of TWT_010 function within the T. whipplei lifecycle and during host infection, potentially revealing new therapeutic targets.