Recombinant Wolbachia sp. subsp. Drosophila simulans Serine hydroxymethyltransferase (glyA)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder. We will preferentially ship the format we have in stock. If you have special format requirements, please note them when ordering, and we will try to accommodate your request.
Lead Time
Delivery times vary depending on the purchasing method and location. Consult your local distributor for specific delivery times. All proteins are shipped with normal blue ice packs by default. For dry ice shipping, please contact us in advance; extra fees apply.
Notes
Avoid repeated freezing and thawing. Working aliquots can be stored at 4°C for up to one week.
Reconstitution
Briefly centrifuge the vial before opening to collect contents at the bottom. 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 default final glycerol concentration is 50%.
Shelf Life
Shelf life depends on several factors: storage state, buffer ingredients, storage temperature, and protein stability. Generally, the liquid form has a shelf life of 6 months at -20°C/-80°C, while the lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during the manufacturing process. If you require a specific tag, please inform us, and we will prioritize developing it.
Synonyms
glyA; WRi_010650Serine hydroxymethyltransferase; SHMT; Serine methylase; EC 2.1.2.1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-425
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Wolbachia sp. subsp. Drosophila simulans (strain wRi)
Target Names
glyA
Target Protein Sequence
MMSVLKKICG SKNSLKSFDN EVYQSIEKEL QRQKSQLQLI ASENFASKAV MEAQGSFLTN KYAEGYPGKR YYCGCEHVDK IESLAIERLC KLFGVKFANV QPHSGSQANQ AVFASLLTPG DTILGLSLSC GGHLTHGAAP SLSGKWFKSI QYTVNKDTYL LDMDEIEKLA LEHKPKLIIA GASAYPRKMD FKRFREIADK VGAYLLADIA HYAGLIAAGE YPSPAEYAHV MTSTTHKTLR GPRGGIVMTN DEILHKKIQS AVFPGLQGGP LMHVIAAKAV AFKEALAPEF KTYSKKVVEN AKVLAQELQK HGLDIITGGT DSHIVLVDLR SQKLTGKDVV DSLERAGITC NKNSVPFDTA KPTITSGLRF GTAAETTRGL EAENFKEIAD LINEVIQGLI SGNSSSVEKA VKAKVERICS NFPIY
Uniprot No.

Target Background

Function
Catalyzes the reversible interconversion of serine and glycine, using tetrahydrofolate (THF) as the one-carbon carrier. This reaction is the primary source of one-carbon groups needed for biosynthesis of purines, thymidylate, methionine, and other essential biomolecules. Also displays THF-independent aldolase activity towards beta-hydroxyamino acids, producing glycine and aldehydes through a retro-aldol mechanism.
Database Links
Protein Families
SHMT family
Subcellular Location
Cytoplasm.

Q&A

How does Wolbachia glyA compare to homologous enzymes in other bacterial species?

Comparative genomic analysis between different Wolbachia strains reveals that while core metabolic genes tend to be conserved, there are often strain-specific adaptations that reflect host specialization. Though specific comparative studies of glyA across Wolbachia strains are not detailed in the available literature, analysis of other metabolic genes suggests that Wolbachia enzymes often display unique characteristics compared to their counterparts in free-living bacteria.

What challenges exist in expressing and purifying recombinant Wolbachia proteins?

Expressing and purifying recombinant proteins from Wolbachia presents significant challenges due to its status as an obligate intracellular bacterium. These challenges include:

  • Limited starting material: As outlined in the literature, researchers typically require large amounts of infected host tissue (traditionally around 10g or thousands of individuals) to obtain sufficient Wolbachia material .

  • Contamination with host DNA: Studies consistently report high levels of host DNA contamination (up to 36% in libraries constructed for wMel sequencing) .

  • Codon usage bias: Adaptation to intracellular lifestyle can result in codon usage patterns that differ significantly from common expression hosts like E. coli.

  • Protein folding and solubility issues: Recombinant expression often results in misfolding or inclusion body formation, requiring optimization of expression conditions.

Researchers have developed strategies to overcome these challenges, including multiple-displacement amplification of Wolbachia DNA from limited starting material (as little as 0.2g tissue or 2×10^7 cells) to obtain 8-10μg of DNA suitable for downstream applications . This approach significantly improves the feasibility of cloning and expressing Wolbachia genes, including glyA.

What is the most efficient protocol for isolating Wolbachia DNA for glyA amplification?

An efficient protocol for isolating Wolbachia DNA suitable for glyA amplification combines differential centrifugation, pulsed-field gel electrophoresis (PFGE), and multiple-displacement amplification. Based on published methodologies, the following procedure is recommended:

  • Initial preparation: Harvest approximately 2×10^7 infected cells (e.g., from Drosophila simulans eggs or infected cell lines like Aa23) .

  • Cell lysis and debris removal:

    • Lyse cells by sonication on ice (90% power, two cycles of 1-minute exposure)

    • Centrifuge at 604×g for 5 minutes to remove cell debris

    • Recover the bacterial fraction from the supernatant

  • Purification and PFGE separation:

    • Prepare the bacterial fraction for PFGE

    • Excise the Wolbachia genome band from the gel

    • Wash the gel slice in TE buffer (four 45-minute washes at 4°C)

    • Digest with gelase according to manufacturer's recommendations

  • DNA amplification:

    • Perform multiple-displacement amplification on the purified DNA

    • Verify amplification by PCR targeting Wolbachia-specific genes

This procedure yields 8-10μg of high-quality Wolbachia DNA from limited starting material, which is sufficient for PCR amplification of specific genes like glyA .

StepTime RequiredCritical ParametersQuality Control
Initial cell preparation1-2 hoursCell viability >90%Trypan blue staining
Cell lysis and fractionation1 hourSonication power and durationMicroscopy check
PFGE separation24-48 hoursVoltage and pulse timeGel imaging
DNA purification5-6 hoursBuffer compositionNanoDrop reading
Multiple-displacement amplification8-16 hoursPrimer design, enzyme qualityPCR verification

How can PCR amplification of glyA from Wolbachia be optimized?

Optimizing PCR amplification of the glyA gene from Wolbachia requires addressing several challenges inherent to this obligate intracellular bacterium. Based on successful approaches for amplifying other Wolbachia genes (such as wsp, dnaJ, gyrB, and cysS), the following optimization strategies are recommended :

  • Primer design considerations:

    • Target conserved regions identified from available Wolbachia genomes

    • Design primers with similar melting temperatures (within 2-3°C)

    • Include appropriate restriction sites for downstream cloning

    • Verify specificity against both Wolbachia and host genomes to prevent non-specific amplification

  • Template preparation:

    • Use DNA isolated and amplified using the multiple-displacement amplification method described in 2.1

    • Determine optimal template concentration (typically 10-50ng per reaction)

    • Assess template quality by amplifying control genes like wsp (Wolbachia surface protein)

  • PCR optimization parameters:

    • Test a range of annealing temperatures (gradient PCR recommended)

    • Optimize MgCl₂ concentration (typically 1.5-3.0mM)

    • Evaluate different polymerases (high-fidelity enzymes recommended for cloning)

    • Add PCR enhancers for GC-rich templates (DMSO, betaine, or specialized buffers)

  • Validation of amplification:

    • Sequence verify PCR products to confirm correct amplification

    • Perform restriction digest analysis to confirm expected fragment patterns

    • Consider cloning and sequencing multiple independent amplicons to detect potential sequence variations

For specific cycling conditions, researchers typically use initial denaturation at 95°C for 5 minutes, followed by 30-35 cycles of denaturation (95°C, 30s), annealing (55-58°C, 30s), and extension (72°C, 90s for a ~1.2kb gene), with a final extension at 72°C for 10 minutes .

What expression systems are most suitable for producing functional recombinant glyA?

The selection of an appropriate expression system for recombinant Wolbachia glyA should consider several factors including protein folding requirements, post-translational modifications, and intended downstream applications. Based on successful expression of other Wolbachia proteins, the following systems can be considered:

  • E. coli-based expression:

    • BL21(DE3) strains are commonly used for initial expression trials

    • Consider codon-optimized constructs to address potential codon bias issues

    • Fusion tags (His6, MBP, GST, SUMO) can improve solubility and facilitate purification

    • Low-temperature induction (16-18°C) often improves proper folding

  • Insect cell expression systems:

    • Sf9 or High Five™ cells with baculovirus vectors

    • More suitable for proteins requiring complex folding or post-translational modifications

    • Closer to the native environment of Wolbachia proteins

    • Longer development time but potentially higher functional yield

  • Cell-free expression systems:

    • Useful for proteins that may be toxic to host cells

    • Allows direct control over reaction conditions

    • Enables incorporation of modified amino acids if needed

    • Limited scale but rapid results for initial characterization

The selection of an optimal expression system should be guided by preliminary small-scale expression trials in multiple systems, with assessment of both yield and functional activity. The functional activity can be determined using enzyme assays specific to serine hydroxymethyltransferase, such as spectrophotometric monitoring of THF conversion or coupled enzyme assays.

How can genomic approaches be applied to study glyA variation across Wolbachia strains?

Genomic approaches provide powerful tools for studying glyA variation across different Wolbachia strains, offering insights into evolutionary adaptations and functional conservation. Based on genomic studies of other Wolbachia genes, the following approaches are recommended:

  • Comparative sequence analysis:

    • Analyze glyA sequences from multiple Wolbachia strains (wMel, wRi, wBm, etc.)

    • Identify conserved domains and variable regions

    • Calculate nucleotide diversity and synonymous/non-synonymous substitution rates

    • Map variations to functional domains based on homology modeling

  • Phylogenetic analysis:

    • Construct phylogenetic trees based on glyA sequences

    • Compare glyA-based phylogenies with those derived from other genes (wsp, MLST genes)

    • Identify potential horizontal gene transfer or recombination events

    • Correlate glyA variants with host species and geographical distribution

  • Population genomics:

    • Analyze glyA variation within populations over time

    • Study selection pressures acting on glyA in different environments

    • Assess the stability of glyA sequences during long-term host association, similar to stability studies performed for wMel in Aedes aegypti populations

  • Integration with other omics data:

    • Correlate glyA variants with transcriptomic profiles

    • Link sequence variations to protein expression levels

    • Associate glyA variants with metabolomic differences between strains

Research on the long-term stability of Wolbachia strains in host populations suggests that core metabolic genes like glyA likely remain relatively stable over time, similar to the genomic stability observed in wMel from field populations collected up to a decade apart . This stability makes glyA a potentially valuable marker for strain identification and evolutionary studies.

What approaches can detect and resolve potential host DNA contamination when studying glyA?

Host DNA contamination represents a significant challenge when studying Wolbachia genes, with studies reporting up to 36% host contamination in genomic libraries . Several approaches can be employed to detect and resolve this issue:

  • Purification optimization:

    • Implement the differential centrifugation and PFGE separation techniques described in section 2.1

    • Use density gradient centrifugation to separate bacterial and host fractions

    • Treat samples with DNase before bacterial lysis to degrade exposed host DNA

  • PCR-based approaches:

    • Design highly specific primers that selectively amplify Wolbachia glyA

    • Implement touchdown PCR protocols to increase specificity

    • Include control reactions with templates from uninfected hosts to identify non-specific amplification

    • Sequence verify all PCR products before downstream applications

  • Bioinformatic screening:

    • When sequencing glyA, compare results against both Wolbachia and host genome databases

    • Implement k-mer based tools to identify potential chimeric sequences

    • Use sequence coverage analysis to identify anomalous regions that may represent contamination

    • Apply phylogenetic placement methods to confirm sequence origin

  • Quantitative assessment:

    • Perform qPCR targeting both Wolbachia glyA and host genes to quantify contamination levels

    • Calculate Wolbachia-to-host DNA ratios at different purification stages

    • Establish minimum purity thresholds for downstream applications

  • Verification strategies:

    • Clone and sequence multiple independent amplicons to ensure consistency

    • Perform restriction fragment length polymorphism (RFLP) analysis using enzymes that differentially cut host and Wolbachia sequences

    • Confirm results using complementary approaches such as RNA-seq data where available

How can functional assays for recombinant glyA be developed and validated?

Developing reliable functional assays for recombinant Wolbachia glyA requires careful consideration of enzyme properties and reaction conditions. The following approaches are recommended based on established methodologies for serine hydroxymethyltransferase characterization:

  • Spectrophotometric assays:

    • Monitor the conversion of tetrahydrofolate (THF) to 5,10-methylene-THF

    • Follow the reaction spectrophotometrically at 340nm using coupled reactions with NADPH

    • Optimize buffer conditions, pH, temperature, and cofactor concentrations

    • Validate with commercially available SHMT from other sources as positive controls

  • Radiochemical assays:

    • Use 14C-labeled serine as substrate

    • Measure conversion to labeled glycine and 5,10-methylene-THF

    • More sensitive than spectrophotometric methods for low activity preparations

    • Requires appropriate radioisotope handling facilities

  • Biochemical characterization:

    • Determine kinetic parameters (Km, Vmax, kcat) for both forward and reverse reactions

    • Assess cofactor requirements (pyridoxal phosphate) and metal ion dependencies

    • Evaluate pH and temperature optima and stability profiles

    • Compare parameters with SHMT enzymes from related organisms

  • Activity validation approaches:

    • Complementation assays in E. coli glyA mutants lacking endogenous SHMT activity

    • Mass spectrometry-based metabolite analysis to track substrate conversion

    • Circular dichroism spectroscopy to confirm proper protein folding

    • Thermal shift assays to assess stability and ligand binding

  • Inhibition studies:

    • Test known SHMT inhibitors to confirm mechanism conservation

    • Develop control reactions with specific inhibitors as negative controls

    • Assess competition with substrate analogs

For all functional assays, appropriate controls should include heat-inactivated enzyme, reaction mixtures lacking individual components, and comparison with commercially available SHMT enzymes. Statistical validation should involve multiple independent preparations and replicate measurements to ensure reproducibility.

How should researchers interpret evolutionary signals in glyA sequences across Wolbachia strains?

Interpretation of evolutionary signals in Wolbachia glyA sequences requires careful consideration of the bacteria's unique lifestyle and genomic characteristics. Based on evolutionary analyses of other Wolbachia genes, researchers should consider:

The interpretation should consider the decade-scale stability observed in some Wolbachia strains , suggesting that core metabolic genes like glyA may show conservation of function despite adaptation to specific hosts.

What statistical approaches are appropriate for analyzing glyA expression and activity data?

Appropriate statistical analysis of glyA expression and activity data ensures reliable interpretation and reproducibility. Based on statistical methods used in Wolbachia research, the following approaches are recommended:

  • Experimental design considerations:

    • Include adequate biological replicates (minimum n=3, preferably n≥5)

    • Plan for technical replicates to assess measurement variation

    • Include appropriate positive and negative controls

    • Consider power analysis to determine sample size requirements

  • Data transformation and normalization:

    • Transform proportional data using logit transformation before statistical analysis

    • Normalize expression data to appropriate reference genes

    • Apply suitable transformations (log, square root) to meet assumptions of parametric tests

    • Consider LOESS normalization for high-throughput datasets

  • Statistical test selection:

    • For comparing expression or activity between groups: general linear models (GLMs)

    • For survival or longevity data: log-rank tests or Cox proportional hazards models

    • For time-course experiments: repeated measures ANOVA or mixed-effects models

    • For complex experimental designs: nested ANOVA with appropriate factors

  • Multiple testing correction:

    • Apply Bonferroni correction when testing multiple traits in the same cohort

    • Consider false discovery rate (FDR) correction for high-throughput data

    • Report both uncorrected and corrected p-values for transparency

    • Establish appropriate significance thresholds based on correction method

  • Data reporting standards:

    • Include measures of central tendency (mean) and dispersion (standard error, confidence intervals)

    • Report exact p-values rather than significance thresholds when possible

    • Provide access to raw data for reanalysis

    • Clearly state all statistical tests and software used

These approaches align with statistical methods used in published Wolbachia research, such as the analysis of phenotypic stability in wMel-infected mosquito populations .

How can researchers distinguish between glyA functional defects and experimental artifacts?

Distinguishing between genuine functional defects in recombinant Wolbachia glyA and experimental artifacts requires systematic troubleshooting and validation approaches:

  • Protein quality assessment:

    • Verify protein integrity using SDS-PAGE and western blotting

    • Confirm protein identity using mass spectrometry

    • Assess oligomeric state using size exclusion chromatography

    • Evaluate protein folding using circular dichroism or fluorescence spectroscopy

  • Cofactor and reaction condition verification:

    • Test enzyme activity with and without added pyridoxal phosphate cofactor

    • Perform activity assays across a range of pH values and temperatures

    • Assess dependence on buffer components and salt concentrations

    • Evaluate potential inhibition by components of the expression or purification system

  • Control experiments:

    • Compare with wild-type enzyme from related species when available

    • Create site-directed mutants of known catalytic residues as negative controls

    • Test activity of the same protein prepared under different conditions

    • Perform parallel assays with commercially available SHMT enzymes

  • Multi-method validation:

    • Compare results from different activity assay methodologies

    • Verify findings using both in vitro and in vivo approaches when possible

    • Apply complementation assays in appropriate bacterial mutants

    • Consider the effects of expression tags and their removal on enzyme function

  • Statistical evaluation:

    • Apply rigorous statistical analysis as outlined in section 4.2

    • Establish clear criteria for distinguishing significant differences

    • Report variability between protein preparations

    • Consider the biological relevance of observed differences

  • Correlation with structural predictions:

    • Map potential defects to structural models based on homology

    • Evaluate whether observed defects align with predicted functional impacts of mutations

    • Consider compensatory mutations that may restore function despite changes

Similar validation approaches have been applied in studies examining the effects of tetracycline treatment on Wolbachia, where researchers carefully distinguished between direct effects on Wolbachia and indirect effects on the host microbiome .

What are the most common pitfalls in Wolbachia protein expression and how can they be avoided?

Expression of Wolbachia proteins, including glyA, presents several challenges that researchers should anticipate and address:

  • Low expression levels:

    • Issue: Codon bias differences between Wolbachia and expression hosts

    • Solution: Use codon-optimized synthetic genes designed for the expression host

    • Validation: Confirm expression using western blot with sensitive detection methods

  • Protein insolubility:

    • Issue: Formation of inclusion bodies during overexpression

    • Solution: Lower induction temperature (16-18°C), reduce inducer concentration, use solubility-enhancing fusion tags (MBP, SUMO)

    • Validation: Compare soluble and insoluble fractions by SDS-PAGE

  • Host toxicity:

    • Issue: Toxic effects of Wolbachia protein expression on host cells

    • Solution: Use tightly controlled inducible systems, consider cell-free expression alternatives

    • Validation: Monitor growth curves following induction, test multiple expression strains

  • Improper folding:

    • Issue: Expressed protein lacks activity due to misfolding

    • Solution: Co-express with chaperones (GroEL/GroES), include appropriate cofactors in growth media

    • Validation: Assess secondary structure using circular dichroism, perform thermal shift assays

  • Protein degradation:

    • Issue: Rapid degradation of expressed protein

    • Solution: Include protease inhibitors, reduce time and temperature during purification steps

    • Validation: Monitor protein stability during storage using activity assays and SDS-PAGE

  • Cofactor incorporation:

    • Issue: Insufficient incorporation of pyridoxal phosphate cofactor

    • Solution: Supplement expression media with pyridoxine, add PLP during purification

    • Validation: Compare UV-visible spectra with and without added cofactor

  • Purification challenges:

    • Issue: Co-purification of host proteins with similar properties

    • Solution: Implement multi-step purification strategies, consider orthogonal purification tags

    • Validation: Assess purity by SDS-PAGE and mass spectrometry

Each of these challenges has been encountered in work with other Wolbachia proteins, and the solutions are based on successful strategies for expressing challenging bacterial proteins from obligate intracellular organisms .

How can researchers verify the purity and identity of recombinant glyA preparations?

Verifying the purity and identity of recombinant Wolbachia glyA requires a combination of analytical techniques:

  • Purity assessment:

    • SDS-PAGE with Coomassie or silver staining (expect >90% purity for kinetic studies)

    • Densitometry analysis to quantify contaminant bands

    • High-resolution techniques such as capillary electrophoresis for higher sensitivity

    • Size exclusion chromatography to detect aggregates and non-specific complexes

  • Identity confirmation:

    • Western blotting with antibodies against the target protein or fusion tags

    • Peptide mass fingerprinting using tryptic digestion and MALDI-TOF

    • Liquid chromatography-tandem mass spectrometry (LC-MS/MS) for sequence coverage

    • N-terminal sequencing to confirm the correct start of the protein

  • Functional verification:

    • Enzymatic activity assays specific to serine hydroxymethyltransferase

    • Cofactor binding analysis (absorption spectrum for PLP-binding)

    • Thermal shift assays to confirm ligand binding capabilities

    • Circular dichroism to verify secondary structure elements

  • Contaminant testing:

    • Endotoxin testing for preparations intended for immunological studies

    • Nucleic acid contamination assessment (A260/A280 ratio, specific nuclease treatments)

    • Host cell protein ELISA for detecting trace contaminants

    • Activity assays for potentially contaminating enzymes

  • Batch consistency verification:

    • Establish acceptance criteria for each analytical method

    • Maintain reference standards from well-characterized batches

    • Document batch-to-batch variation for critical quality attributes

    • Implement statistical process control for monitoring trends

These verification methods align with approaches used for other recombinant proteins from challenging sources like Wolbachia, where purity and identity confirmation are essential for reliable functional studies .

What controls should be included when studying glyA function across different experimental conditions?

Robust experimental design for studying Wolbachia glyA function requires carefully selected controls to ensure valid interpretation of results:

  • Positive and negative controls:

    • Positive control: Commercial or well-characterized SHMT from another organism

    • Negative control: Heat-inactivated enzyme, catalytic site mutant, or reaction missing essential components

    • System control: Expression and purification of a known functional protein using identical methods

  • Experimental condition controls:

    • Include buffer-only controls for all experimental conditions

    • Test vehicle controls for all additives (solvents, stabilizers)

    • Create condition matrices to identify interaction effects

    • Include time-zero measurements for all kinetic studies

  • Biological reference standards:

    • When comparing glyA variants, include a well-characterized reference standard in each experiment

    • Maintain consistent positive control sources across studies

    • Include internal normalization controls for relative activity measurements

    • Document passage number or preparation date for all biological materials

  • Methodological controls:

    • Test multiple detection methods when possible

    • Include standard curves with each quantitative assay

    • Perform spike recovery experiments to assess matrix effects

    • Run parallel assays with alternative substrates or cofactors

  • Controls for specificity:

    • Include closely related enzymes to confirm assay specificity

    • Test potential inhibitors with known mechanisms

    • Include competition assays with substrate analogs

    • Verify substrate identity before and after reaction using analytical methods

  • Replicate controls:

    • Technical replicates to assess method precision

    • Biological replicates to capture natural variation

    • Independent experimental repeats on different days

    • Inter-operator reproducibility tests for complex protocols

Implementing these controls helps distinguish genuine biological effects from experimental artifacts, a critical consideration when working with challenging proteins like those from Wolbachia. The approach follows established practices in enzyme characterization while addressing the specific challenges of working with recombinant proteins from obligate intracellular bacteria .

What are the most promising future directions for Wolbachia glyA research?

Research on Wolbachia sp. subsp. Drosophila simulans Serine hydroxymethyltransferase (glyA) has several promising future directions that build upon current methodologies and findings:

  • Comparative genomics and evolution:

    • Expanded analysis of glyA across multiple Wolbachia strains from diverse hosts

    • Investigation of selection pressures on glyA in different host environments

    • Long-term evolutionary studies similar to the decade-long stability assessment of wMel

    • Integration of glyA variation data with whole-genome phylogenetic analyses

  • Structure-function relationships:

    • Determination of three-dimensional structure through X-ray crystallography or cryo-EM

    • Structure-guided mutagenesis to identify host-specific adaptations

    • Comparative structural biology with homologous enzymes from free-living bacteria

    • Computational modeling of enzyme dynamics and substrate interactions

  • Metabolic integration studies:

    • Investigation of glyA's role in Wolbachia-host metabolic interactions

    • Metabolomic profiling to trace serine/glycine/folate metabolism in infected versus uninfected hosts

    • Integration of glyA function into systems biology models of Wolbachia metabolism

    • Exploration of potential metabolic complementation between Wolbachia and host pathways

  • Applied research directions:

    • Development of glyA-targeted approaches for manipulating Wolbachia infections

    • Investigation of glyA as a potential drug target for anti-filarial applications

    • Exploration of glyA's role in phenotypes like cytoplasmic incompatibility

    • Assessment of glyA as a marker for Wolbachia strain typing and population studies

  • Methodological advances:

    • Refinement of purification techniques to improve yields from limited starting material

    • Development of high-throughput screening methods for glyA inhibitors

    • Creation of conditional expression systems for studying glyA function in vivo

    • Implementation of CRISPR-based approaches for manipulating glyA in situ

These research directions build upon the foundation of successful Wolbachia research methodologies demonstrated in long-term studies like the decade-long assessment of wMel stability in Aedes aegypti and the efficient DNA purification methods developed for Wolbachia genomic studies .

How might glyA research contribute to our understanding of Wolbachia-host interactions?

Research on Wolbachia glyA has significant potential to advance our understanding of Wolbachia-host interactions across multiple dimensions:

  • Metabolic integration understanding:

    • Clarification of how Wolbachia supplements or complements host metabolism

    • Insights into nutritional dependencies between Wolbachia and its host

    • Understanding of metabolic changes during Wolbachia adaptation to new hosts

    • Potential parallels to the "mitochondrion-like function" described for Wolbachia in generating ATP for hosts

  • Host manipulation mechanisms:

    • Investigation of glyA's potential role in phenotypes like cytoplasmic incompatibility

    • Examination of how one-carbon metabolism influences host reproduction

    • Study of metabolic competition or cooperation between Wolbachia and other symbionts

    • Potential influences on host immune responses, similar to the immunogenic proteins identified in wOo

  • Evolutionary perspectives:

    • Analysis of how glyA adaptation reflects Wolbachia's co-evolution with hosts

    • Insights into metabolic gene retention during genome streamlining

    • Understanding of selection pressures in different host environments

    • Potential correlations between glyA variants and phenotypic effects in hosts

  • Applied potential:

    • Development of metabolism-based strategies for manipulating Wolbachia

    • Insights into molecular mechanisms of successful Wolbachia-based interventions

    • Understanding of metabolic factors affecting Wolbachia stability in novel hosts

    • Identification of potential targets for controlling Wolbachia-mediated phenotypes

  • Host range determinants:

    • Investigation of how metabolic complementation influences host range

    • Analysis of glyA adaptations in strains with different host specificities

    • Correlation between metabolic capabilities and successful host colonization

    • Understanding factors contributing to the long-term stability observed in established Wolbachia infections

This research would complement existing knowledge about Wolbachia's genome, transcriptome, and proteome, providing a more comprehensive understanding of the metabolic dimensions of this important symbiotic relationship .

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.