Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes. We will accommodate your request if possible.
Note: All protein shipments include standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
AIM4 (Altered Inheritance of Mitochondria protein 4) in C. glabrata is involved in mitochondrial genome maintenance and proper mitochondrial distribution during cell division. The protein contributes to mitochondrial DNA stability and plays a critical role in energy metabolism. While research specifically on C. glabrata AIM4 is still evolving, studies suggest it functions similarly to homologous proteins in related fungal species. To effectively study AIM4 function, researchers typically employ gene deletion strategies followed by phenotypic characterization of respiratory capacity, mitochondrial morphology visualization using fluorescent markers, and stress tolerance assays. Similar to studies on other C. glabrata proteins, research methodologies might parallel those used for investigating proteins like CgDtr1, which has been identified as a determinant of virulence .
Comparative analysis of AIM4 across fungal species reveals conserved domains that suggest evolutionary preservation of core functions in mitochondrial maintenance. When analyzing AIM4 proteins, researchers should:
Perform sequence alignment using tools like MUSCLE or Clustal Omega across fungal species
Identify conserved domains using Pfam or SMART databases
Generate phylogenetic trees to establish evolutionary relationships
Use structural prediction tools like I-TASSER or AlphaFold to predict protein structure
Despite C. glabrata's closer phylogenetic relationship to Saccharomyces cerevisiae than to Candida albicans, functional comparative studies should consider that homologous recombination works with poor efficiency in C. glabrata compared to baker's yeast . This genetic engineering limitation should be factored into experimental design when planning comparative functional studies.
Given the poor homologous recombination efficiency in C. glabrata, researchers should consider using DNA Ligase IV (LIG4) deletion strains to improve targeted genetic manipulations. The table below compares key genetic manipulation techniques:
| Technique | Efficiency in C. glabrata | Advantages | Limitations |
|---|---|---|---|
| Standard homologous recombination | Very low (~0.2%) | Simple methodology | Extremely inefficient, many off-target integrations |
| LIG4 deletion background | Significantly improved | Specific for gene targeting, minimal side effects | Requires initial generation of LIG4 mutant strain |
| KU80 deletion background | Improved | Increases homologous recombination | May have unwanted side effects on general DNA repair |
| CRISPR-Cas9 | High | Precise genome editing | Requires optimization for C. glabrata |
The LIG4 deletion approach has been demonstrated to significantly increase correct gene targeting without detectable side effects on growth, DNA stress tolerance, or antifungal drug resistance . When designing gene deletion constructs for AIM4, use 500-bp promoter and terminator sequences flanking a selectable marker gene (like ScHIS3) as demonstrated effective in previous C. glabrata genetic studies .
To accurately measure AIM4 expression and confirm its mitochondrial localization:
Expression quantification:
Develop C-terminal or N-terminal epitope-tagged versions of AIM4 (consider HA, FLAG or GFP tags)
Use qRT-PCR to measure transcript levels under different conditions
Employ Western blotting with specific antibodies to quantify protein levels
Consider mass spectrometry-based proteomics for precise quantification
Localization studies:
Fluorescence microscopy using GFP-tagged AIM4 co-stained with mitochondrial markers (MitoTracker)
Subcellular fractionation followed by Western blot analysis
Immunoelectron microscopy for high-resolution localization
Proximity labeling methods such as BioID or APEX to identify proximal interacting proteins
When designing these experiments, consider that mitochondrial proteins often require specific targeting sequences for proper localization. Mutations in these sequences may affect localization patterns, similar to how mutations in the CgDtr1 transporter affect its plasma membrane localization and function in stress resistance .
Based on successful approaches with other C. glabrata virulence factors, researchers should employ a multi-faceted strategy:
In vitro infection models:
Galleria mellonella larval infection assay: Inject ~5 × 10^7 CFU/larvae of wild-type and AIM4 deletion mutants, then monitor survival over 72 hours using Kaplan-Meier survival curves
Cell culture models with hemocytes or macrophages: Compare internalization and proliferation rates between wild-type and mutant strains
Quantitative analysis of fungal burden in host tissues at multiple time points (1h, 24h, 48h) to assess proliferation capacity
Stress tolerance assays:
Oxidative stress (H₂O₂, menadione)
Acetic acid tolerance (particularly relevant as acid stress occurs within phagosomes)
Antimicrobial peptide resistance tests
Antifungal drug susceptibility testing
Virulence gene expression:
RNA-seq analysis comparing wild-type and AIM4 mutants under infection-mimicking conditions
ChIP-seq to identify potential transcriptional regulatory roles
When analyzing virulence results, remember that some C. glabrata strains show inherent differences in virulence capacity. For example, the L5U1 wild-type strain appears less virulent than the KUE100 wild-type strain in Galleria mellonella infection models , highlighting the importance of using appropriate controls and considering strain background effects.
To assess AIM4 function under mitochondrial stress conditions:
Stress induction protocols:
Treat cells with respiratory chain inhibitors (antimycin A, oligomycin)
Apply oxidative stress agents (H₂O₂, paraquat)
Grow cells under hypoxic conditions
Expose to mitochondrial DNA damaging agents
Analytical approaches:
Measure mitochondrial membrane potential using fluorescent dyes (JC-1, TMRM)
Assess mitochondrial DNA stability through qPCR-based mtDNA quantification
Monitor mitochondrial morphology changes via fluorescence microscopy
Measure respiratory capacity using oxygen consumption rate assays
Analyze mitochondrial protein import efficiency
Inheritance pattern analysis:
Time-lapse microscopy to track mitochondrial distribution during cell division
Single-cell analysis of mtDNA copy number variation
Mathematical modeling of mitochondrial segregation patterns
When designing these experiments, consider that mitochondrial inheritance patterns show similarities to those observed in human mitochondrial DNA transmission, where bottleneck effects and selection pressures influence the persistence of mutations . This can provide valuable comparative insights for your research.
When analyzing experimental data related to AIM4 function:
For growth and stress tolerance assays:
Use two-way ANOVA with Bonferroni post-hoc tests to compare wild-type and mutant strains across multiple conditions
Apply regression analysis for dose-response relationships to stress agents
Consider time-series analysis for growth dynamics
For microscopy-based localization data:
Employ colocalization coefficients (Pearson's, Mander's)
Use automated image analysis algorithms for unbiased quantification
Consider machine learning approaches for pattern recognition in complex images
For virulence studies:
Apply log-rank tests for survival curve comparisons
Use appropriate non-parametric tests for CFU quantification from infection models
Include power calculations to ensure adequate sample sizes
When reporting statistical significance, follow the conventions established in the literature: * P < 0.05; ** P < 0.01; *** P < 0.001 , and ensure biological replicates (n≥3) for all critical experiments.
To differentiate direct from indirect effects:
Complementation strategies:
Reintroduce wild-type AIM4 to confirm phenotype rescue
Use point mutants affecting specific domains to identify critical functional regions
Employ heterologous expression of AIM4 homologs from related species
Temporal analysis:
Utilize inducible promoter systems to control AIM4 expression
Perform time-course experiments to determine primary vs. secondary effects
Apply metabolic flux analysis to track changes in mitochondrial function over time
Interaction studies:
Conduct synthetic genetic array analysis to identify genetic interactions
Perform protein-protein interaction studies (co-IP, Y2H, BioID)
Use metabolomics to identify altered metabolic pathways
Controlled expression systems:
For successful purification of recombinant AIM4:
Expression systems:
E. coli: BL21(DE3) with codon optimization for mitochondrial proteins
Yeast: P. pastoris or S. cerevisiae for eukaryotic processing
Insect cell/baculovirus system for complex proteins
Cell-free expression systems for potentially toxic proteins
Purification strategies:
Affinity tags: His6, GST, MBP (consider tag position effects)
Size exclusion chromatography for oligomeric state determination
Ion exchange chromatography for charged variants separation
Native vs. denaturing conditions optimization
Protein quality assessment:
Circular dichroism for secondary structure analysis
Thermal shift assays for stability assessment
Dynamic light scattering for aggregation analysis
Limited proteolysis to identify stable domains
| Expression System | Advantages | Limitations | Best for |
|---|---|---|---|
| E. coli | High yield, low cost | Limited post-translational modifications | Soluble domains, protein fragments |
| S. cerevisiae | Eukaryotic processing, genetic tools | Moderate yield | Full-length protein with proper folding |
| Insect cells | High-quality eukaryotic processing | Higher cost, longer timeline | Complex proteins requiring chaperones |
| Cell-free | Rapid, good for toxic proteins | Lower yield, higher cost | Initial screening, toxic proteins |
When designing experiments to study AIM4 mutations:
Essential controls:
Wild-type strain (parental background)
Complete gene deletion mutant
Complemented strain with wild-type AIM4
Empty vector controls for all plasmid-based experiments
Positive controls for mitochondrial inheritance defects (e.g., known mutants)
Experimental validation:
Test multiple independent clones to rule out clone-specific effects
Confirm genotypes by PCR and sequencing
Verify protein expression/absence by Western blot
Include growth conditions that don't require mitochondrial function (fermentable carbon sources)
Phenotypic analysis framework:
Categorical classification of mitochondrial morphology patterns
Quantitative measurements of mtDNA copy number
Assessment of mitochondrial distribution symmetry during cell division
Analysis across multiple growth conditions and stress states
When analyzing mitochondrial inheritance patterns, remember that bottleneck effects similar to those observed in human mitochondrial DNA transmission may influence the interpretation of results, especially in clonal populations.
Understanding AIM4's role in mitochondrial function provides several potential therapeutic approaches:
Target identification strategies:
Identify structural differences between fungal and human mitochondrial proteins
Screen for small molecules that specifically inhibit C. glabrata AIM4
Explore the AIM4 interactome for additional targetable proteins
Consider combination approaches targeting multiple mitochondrial functions
Therapeutic development approaches:
Structure-based drug design targeting AIM4-specific pockets
Peptide inhibitors mimicking critical interaction domains
RNA interference or antisense oligonucleotides for gene silencing
Mitochondrial-targeted drug delivery systems
Resistance management considerations:
Assess frequency of spontaneous resistance
Characterize cross-resistance patterns with existing antifungals
Develop combination therapy approaches
Monitor for compensatory mechanisms after AIM4 inhibition
Research into mitochondrial proteins as antifungal targets should consider the clinical relevance of C. glabrata as the second most prevalent human opportunistic fungal pathogen in the United States, with increasing incidence and inherent tolerance toward commonly used azole antifungal drugs .
To evaluate AIM4 as a potential diagnostic biomarker:
Expression analysis in clinical settings:
Compare AIM4 expression levels between laboratory and clinical isolates
Assess expression changes during infection progression
Determine if AIM4 is secreted or released during infection
Evaluate expression in drug-resistant vs. susceptible strains
Diagnostic development pipeline:
Generate specific antibodies against unique AIM4 epitopes
Develop ELISA or lateral flow assays for protein detection
Explore aptamer-based detection methods
Consider PCR-based detection of AIM4 genomic sequences
Validation approaches:
Test with diverse clinical isolates to assess conservation
Determine specificity against other Candida species and common pathogens
Establish sensitivity limits in relevant biological matrices
Conduct time-course studies to determine optimal sampling timing
Clinical correlation analysis:
Assess relationship between AIM4 levels and disease severity
Correlate with treatment response
Evaluate prognostic value
Compare with existing diagnostic methods
As with other virulence factors in C. glabrata, such as CgDtr1 , the relationship between AIM4 expression and pathogenesis should be thoroughly characterized before proceeding with biomarker development efforts.