Recombinant Candida glabrata Sorting Nexin-4 (SNX4)
Sorting nexin 4 (SNX4) is involved in vacuole separation and division throughout the cell cycle. Its functions include the retrieval of late-Golgi SNAREs from post-Golgi endosomes to the trans-Golgi network, and it plays a role in cytoplasm-to-vacuole transport (Cvt), autophagy, mitophagy, and pexophagy.
KEGG: cgr:CAGL0J01001g
STRING: 284593.XP_447755.1
Sorting nexin-4 in C. glabrata likely contains the characteristic domains found in other SNX family proteins, including a phosphatidylinositol 3-phosphate binding domain (phox homology or PX domain) necessary for peripheral membrane localization, and a C-terminal Bin/Amphiphysin/Rvs (BAR) domain that recognizes and binds to curved membranes upon dimerization . The PX domain specifically binds to phosphatidylinositol 3-phosphate, recruiting SNX4 to endosomal membranes . Like other SNX-BAR proteins, C. glabrata SNX4 likely forms homodimers or heterodimers with other sorting nexins to create membrane tubules that facilitate protein trafficking between endosomal compartments and the plasma membrane . Based on conservation with other fungal species, C. glabrata SNX4 would be expected to participate in recycling pathways that prevent cargo degradation in the vacuole (the fungal equivalent of lysosomes) .
When designing a recombinant expression system for C. glabrata SNX4, several factors should be considered:
Expression host selection: While E. coli provides high yield and rapid growth, yeast expression systems like Saccharomyces cerevisiae may offer better protein folding due to their closer evolutionary relationship to C. glabrata. Since proper folding of both the PX and BAR domains is critical for SNX4 function, a eukaryotic expression system may be preferable for functional studies.
Affinity tags: Include an N- or C-terminal tag (His6, GST, MBP) to facilitate purification while ensuring that the tag doesn't interfere with membrane binding or protein interactions. C-terminal tags may be preferable if the N-terminus contains functional motifs.
Solubility considerations: SNX4 is a membrane-associated protein, so expression conditions should be optimized to prevent aggregation. Consider fusion partners that enhance solubility (MBP, SUMO) and include appropriate detergents in purification buffers.
Domain-based constructs: For structural studies, express individual domains (PX domain, BAR domain) separately, as full-length protein may exhibit conformational flexibility that complicates crystallization.
For optimal purification of recombinant C. glabrata SNX4:
Lysis buffer composition:
50 mM Tris or HEPES pH 7.5-8.0
300 mM NaCl (adjust based on solubility)
5-10% glycerol for stability
1-5 mM DTT or TCEP as reducing agent
Protease inhibitor cocktail
Consider mild detergents (0.1% Triton X-100 or 1% CHAPS) if solubility is an issue
Multi-step purification strategy:
Initial capture: Affinity chromatography using tag (Ni-NTA for His-tagged protein)
Intermediate: Ion exchange chromatography to remove contaminants
Final polishing: Size exclusion chromatography to ensure homogeneity and proper oligomeric state
Quality control assessments:
SDS-PAGE and Western blotting to confirm identity and purity
Dynamic light scattering to check for aggregation
Circular dichroism to verify proper folding
Functional assays (lipid binding) to confirm activity
For membrane binding studies, incorporate liposomes containing phosphatidylinositol 3-phosphate during purification to stabilize the protein in its membrane-bound state.
Expression yields for recombinant C. glabrata SNX4 vary significantly depending on the expression system used:
| Expression System | Expected Yield (mg/L culture) | Advantages | Limitations |
|---|---|---|---|
| E. coli | 5-20 mg/L | High yield, rapid growth | Potential misfolding |
| S. cerevisiae | 2-10 mg/L | Natural folding, PTMs | Longer growth time |
| Pichia pastoris | 10-50 mg/L | High density culture | Complex protocols |
| Insect cells | 1-5 mg/L | Complex folding | Expensive, low yield |
Yields can be improved by optimizing:
Growth temperature (often lower temperatures improve folding)
Induction conditions (concentration of inducer, timing)
Media composition (rich vs. defined media)
Cell lysis methods (gentle extraction preserves structure)
For membrane-associated proteins like SNX4, final yields of active protein are typically lower than cytosolic proteins due to challenges with proper folding and maintaining solubility.
To evaluate the membrane tubulation activity of recombinant C. glabrata SNX4:
In vitro liposome tubulation assay:
Prepare liposomes with composition mimicking endosomal membranes (including PI3P)
Incubate purified SNX4 with liposomes at varying concentrations
Visualize using negative-stain electron microscopy or cryo-EM
Quantify tubule formation by measuring tubule length, diameter, and frequency
GUV-based assays (Giant Unilamellar Vesicles):
Generate GUVs containing fluorescent lipids and PI3P
Add fluorescently labeled SNX4
Observe membrane deformation using confocal microscopy
Quantify curvature generation in real-time
Cellular assays:
Express fluorescently tagged SNX4 in cultured cells
Visualize endosomal tubule formation using live-cell imaging
Quantify tubule dynamics (extension rate, lifetime, frequency)
Compare with known tubulation-defective mutants
Structure-function analysis:
Generate mutations in the BAR domain that disrupt dimerization
Test tubulation activity of mutants vs. wild-type protein
Create chimeric proteins with BAR domains from other sorting nexins
When analyzing results, consider that SNX4 may require additional factors for optimal tubulation activity in vivo, such as interaction partners that are absent in reconstituted systems .
To determine the cargo specificity of C. glabrata SNX4:
Proteomic approaches:
Compare plasma membrane proteomes of wild-type vs. SNX4 knockout strains
Perform SILAC labeling for quantitative comparison
Focus on proteins showing decreased surface expression in knockout
Validate top candidates with targeted approaches
Proximity labeling techniques:
Express SNX4 fused to BioID or APEX2 in C. glabrata
Allow in vivo biotinylation of proteins in proximity to SNX4
Purify biotinylated proteins and identify by mass spectrometry
Compare to control strains expressing only the labeling enzyme
Direct binding assays:
Express cytoplasmic domains of potential cargo proteins
Perform pull-down assays with purified SNX4
Use surface plasmon resonance to measure binding affinities
Map binding interfaces using truncation mutants
Cargo trafficking assays:
Generate fluorescent protein fusions with candidate cargoes
Compare trafficking dynamics in wild-type vs. SNX4 knockout cells
Quantify surface/internal distribution using flow cytometry
Measure protein half-life with cycloheximide chase experiments
When interpreting results, remember that in neurons, SNX4 depletion affects a variety of synaptic membrane proteins rather than just the canonical transferrin receptor cargo seen in other cell types, suggesting context-dependent cargo specificity .
Comparative analysis of C. glabrata SNX4 with homologs in other fungi reveals insights into functional conservation and specialization:
Evolutionary conservation patterns:
The core PX and BAR domains show high sequence conservation across fungal species
Species-specific variations are often found in linker regions and terminal segments
C. glabrata SNX4 likely maintains the fundamental endosomal recycling function
Complementation studies:
In S. cerevisiae, SNX4 (Snx4p) prevents degradation of proteins like the exocytic v-SNARE Snc1p by recycling them from endosomes to the plasma membrane
Cross-species complementation experiments can test if C. glabrata SNX4 rescues phenotypes in S. cerevisiae snx4Δ strains
Failure to complement would suggest species-specific adaptations
Interactome differences:
Cargo specificity variations:
Pathogenic fungi may utilize SNX4 to traffic virulence factors or stress response proteins
Differences in cargo recognition could reflect adaptation to host environments
Comparison with C. albicans SNX4 could reveal pathogen-specific functions
Studies in neurons have shown that SNX4's role may extend beyond the canonical trafficking of transferrin receptor seen in non-neuronal cells, suggesting context-dependent functions that may also exist in different fungal species .
SNX4's potential contributions to C. glabrata pathogenicity and antifungal resistance include:
Regulation of virulence factor trafficking:
C. glabrata SNX4 could control the surface exposure of adhesins needed for host cell attachment
Secretion of hydrolytic enzymes might depend on SNX4-mediated sorting
Immune evasion factors may require SNX4 for proper localization
Stress response adaptation:
C. glabrata thrives in diverse host niches with varying pH, nutrient availability, and immune pressures
SNX4 could regulate the trafficking of stress sensors and response proteins
Rapid membrane composition remodeling during stress might involve SNX4 pathways
Antifungal drug resistance mechanisms:
Adaptation to nutrient limitation:
Experimental approaches to test these hypotheses would include generating C. glabrata SNX4 knockout strains and assessing their virulence in infection models, antifungal susceptibility, and ability to adapt to various stressors.
When analyzing protein trafficking in C. glabrata SNX4 mutants, the following controls are essential:
Genetic validation controls:
Complementation with wild-type SNX4 to confirm phenotype reversal
Multiple independent SNX4 mutant clones to rule out off-target effects
Domain-specific mutants (PX domain, BAR domain) to dissect functions
Empty vector controls for complementation experiments
Cargo protein controls:
Known SNX4-independent cargo proteins as negative controls
Known SNX4-dependent cargo (if identified) as positive controls
Total protein level measurements to distinguish trafficking from expression changes
Cellular localization markers to verify compartment identity
Technical controls:
Microscopy: Proper channel alignment, bleed-through controls, threshold consistency
Biochemical fractionation: Marker proteins for each compartment, loading controls
Flow cytometry: Unstained controls, single-color controls, isotype controls
Image analysis: Randomized blind analysis, consistent ROI selection
Physiological state controls:
Growth phase standardization (log phase cultures)
Media composition consistency
Temperature and pH maintenance
Stress exposure timing for stress response studies
Studies in neurons have shown that SNX4 knockdown effects can vary between specific short hairpin RNAs, suggesting possible off-target effects that must be controlled for by using multiple independent approaches to disrupt SNX4 function .
To troubleshoot protein aggregation issues with recombinant C. glabrata SNX4:
Expression optimization:
Lower induction temperature (16-20°C) to slow folding and prevent aggregation
Reduce inducer concentration for more gradual expression
Shorten induction time to prevent accumulation of misfolded protein
Co-express with molecular chaperones (GroEL/ES, DnaK/DnaJ/GrpE)
Buffer optimization:
Screen buffer compositions systematically (pH 6.0-8.5, NaCl 100-500 mM)
Test different buffering agents (Tris, HEPES, phosphate)
Add stabilizing agents (10% glycerol, 100-500 mM arginine, 1 M urea)
Include reducing agents (5 mM DTT or TCEP) to prevent disulfide-mediated aggregation
Solubility enhancement strategies:
Express as fusion with highly soluble partners (MBP, SUMO, Trx)
Include mild detergents during purification (0.03% DDM, 0.1% CHAPS)
Add PIP3 headgroups or liposomes to stabilize the PX domain
Try extraction with increased salt concentration (500 mM NaCl)
Protein engineering approaches:
Express individual domains separately
Remove flexible regions identified by disorder prediction
Introduce surface mutations to increase solubility
Create truncated constructs based on limited proteolysis results
Systematic screening can be documented in a table format:
| Condition | Temperature | Buffer | Additives | Result |
|---|---|---|---|---|
| 1 | 18°C | 50 mM Tris pH 7.5, 150 mM NaCl | None | Moderate aggregation |
| 2 | 18°C | 50 mM Tris pH 7.5, 150 mM NaCl | 10% glycerol | Improved solubility |
| 3 | 18°C | 50 mM HEPES pH 7.5, 300 mM NaCl | 10% glycerol, 1 mM DTT | Best solubility |
This methodical approach helps identify optimal conditions for obtaining soluble, functional protein.
To resolve contradictory results between in vitro and in vivo studies of C. glabrata SNX4:
Bridging experimental systems:
Semi-permeabilized cell assays that maintain cytosolic factors while allowing controlled addition of recombinant proteins
Cell extract supplementation experiments to identify missing cofactors
Liposome recruitment assays using native membranes isolated from C. glabrata
Reconstitution of purified components in increasing complexity
Protein state verification:
Confirm post-translational modification status of native vs. recombinant SNX4
Validate protein folding using spectroscopic methods (CD, fluorescence)
Check oligomerization state using size exclusion chromatography
Verify membrane binding capacity with liposome flotation assays
Controlled variable testing:
Systematically vary buffer conditions to mimic intracellular environment
Test pH dependence across physiological range (pH 5.5-7.5)
Examine temperature sensitivity of interactions
Assess effects of molecular crowding agents
Interaction partner identification:
Perform pull-downs from C. glabrata lysates using recombinant SNX4
Test if adding specific binding partners restores activity in vitro
Use proximity labeling in vivo to identify the complete SNX4 interactome
Create minimal reconstituted systems with key identified partners
In neurons, SNX4 depletion affects numerous synaptic proteins rather than just the canonical transferrin receptor affected in non-neuronal cells, suggesting context-dependent functions that might explain discrepancies between different experimental systems .
Designing a CRISPR-Cas9 system for modifying SNX4 in C. glabrata requires specific considerations for this pathogenic yeast:
Guide RNA design:
Select target sequences with minimal off-target potential using C. glabrata genome database
Design gRNAs targeting early exons to ensure complete loss of function
Verify PAM sites (NGG for SpCas9) accessibility in the genomic region
Create multiple gRNAs targeting different regions to improve success rates
Repair template construction:
For gene deletion: Design homology arms 500-1000 bp flanking the SNX4 coding region
For epitope tagging: Insert tag sequence in-frame at N- or C-terminus with ~50 bp homology arms
For point mutations: Include the desired mutation with ~50 bp homology on each side
Include selectable markers appropriate for C. glabrata (NAT1, HygB)
Delivery system optimization:
Use lithium acetate/PEG transformation protocol optimized for C. glabrata
Consider electroporation for higher transformation efficiency
Deliver Cas9 and gRNA as ribonucleoprotein complex for transient expression
Use a C. glabrata-optimized Cas9 expression cassette for stable expression
Screening and validation:
Design PCR primers spanning the modification site
Sequence verify all modifications
Confirm protein expression changes by Western blot
Assess potential off-target effects at predicted sites
Example gRNA design parameters:
Target sequence: 20 nucleotides upstream of PAM (NGG)
GC content: 40-60% for optimal binding
Avoid sequences with homology elsewhere in the genome
Target conserved domains for functional disruption
To analyze colocalization of C. glabrata SNX4 with endosomal markers:
Image acquisition considerations:
Use confocal microscopy with appropriate filter sets to minimize bleed-through
Acquire images at Nyquist sampling rate for optimal resolution
Collect Z-stacks to capture the full 3D volume of cells
Include single-labeled controls to correct for spectral overlap
Quantitative colocalization metrics:
Pearson's correlation coefficient (PCC): Measures linear correlation between intensities (-1 to +1)
Manders' overlap coefficients (MOC): Proportion of each signal overlapping with the other (0 to 1)
Object-based methods: Count discrete structures that contain both markers
Analysis workflow:
Apply appropriate background subtraction
Set consistent thresholds across samples
Apply deconvolution if necessary to improve signal-to-noise
Generate scatterplots of pixel intensities from both channels
Calculate coefficients and statistical significance
Interpretation guidelines:
PCC > 0.5 suggests meaningful colocalization
Compare experimental values to randomized controls
Analyze multiple cells (>20) across independent experiments
Consider partial colocalization biologically significant
Studies in neurons have shown that endogenous SNX4 colocalizes with both early endosome marker RAB5 (Pearson's coefficient 0.58) and recycling endosome marker RAB11 (Pearson's coefficient 0.45) . Similar approaches can be applied to C. glabrata studies, with appropriate controls to account for the smaller cell size.
For analyzing SNX4-dependent protein trafficking changes:
Experimental design considerations:
Include biological replicates (minimum n=3) with multiple technical replicates
Use time-course measurements for trafficking kinetics
Include appropriate wild-type and negative controls
Power analysis to determine sample size needed for statistical significance
Statistical tests for different data types:
Normally distributed continuous data: t-test (two conditions) or ANOVA (multiple conditions)
Non-normally distributed data: Mann-Whitney U or Kruskal-Wallis tests
Time-course data: Repeated measures ANOVA or mixed-effects models
Colocalization coefficients: Fisher's z-transformation before parametric testing
Multiple hypothesis testing correction:
Bonferroni correction for stringent control of false positives
Benjamini-Hochberg procedure for false discovery rate control
q-value calculation for large-scale proteomics data
Data visualization approaches:
Box plots showing distribution, median, and outliers
Bar graphs with individual data points visible
Time-course plots with error bars
Heat maps for multiple protein/condition comparisons
Example statistical reporting for trafficking data:
| Protein | Condition | Surface/Total Ratio | p-value | Adjusted p-value |
|---|---|---|---|---|
| Protein A | WT | 0.65 ± 0.08 | - | - |
| Protein A | ΔSNX4 | 0.38 ± 0.07 | 0.0023 | 0.0115 |
| Protein B | WT | 0.42 ± 0.05 | - | - |
| Protein B | ΔSNX4 | 0.45 ± 0.06 | 0.3821 | 0.9552 |
In neurons, quantitative mass spectrometry revealed that upon SNX4 knockdown, proteins involved in neurotransmission were the most dysregulated class . Similar approaches could be applied to C. glabrata to identify trafficking changes systematically.
To interpret proteomic data from C. glabrata SNX4 mutants:
Data processing and normalization:
Apply appropriate normalization methods (global median, spike-in controls)
Log-transform data to approximate normal distribution
Filter low-confidence identifications
Consider batch effects and technical variations
Differential expression analysis:
Calculate fold changes between SNX4 mutant and wild-type
Apply statistical tests with multiple hypothesis correction
Set significance thresholds (typically fold change ≥1.5, p-value <0.05)
Generate volcano plots to visualize significance vs. magnitude
Functional categorization:
Perform Gene Ontology enrichment analysis
Apply pathway analysis (KEGG, Reactome)
Identify protein domains enriched in affected proteins
Analyze cellular compartment enrichment
Biological interpretation:
Focus on membrane proteins that may be direct SNX4 cargoes
Look for changes in known trafficking machinery components
Consider secondary effects due to altered cell physiology
Compare with SNX4 studies in other organisms
In neurons, SNX4 knockdown affected membrane proteins at both presynaptic and postsynaptic terminals involved in processes such as synapse assembly, neurotransmission, and synaptic vesicle cycling . For C. glabrata, focus on membrane proteins involved in stress response, nutrient acquisition, and cell wall maintenance that might be directly affected by SNX4-mediated trafficking.
Research on C. glabrata SNX4 could inform novel antifungal development strategies through several mechanisms:
Targeting SNX4-dependent trafficking pathways:
If SNX4 is essential for C. glabrata virulence or stress response
If SNX4 regulates drug efflux pump localization
If SNX4 mediates cell wall integrity maintenance
Potential drug development approaches:
Small molecule inhibitors targeting the PX domain-phosphoinositide interaction
Peptide-based disruptors of SNX4 protein-protein interactions
Compounds affecting SNX4 dimerization or membrane tubulation
Combinatorial therapy strategies:
SNX4 inhibition to sensitize C. glabrata to existing antifungals
Dual targeting of complementary trafficking pathways
Disruption of SNX4-dependent stress responses combined with stress-inducing antifungals
Structure-based drug design opportunities:
Exploit structural differences between fungal and human SNX4
Target species-specific interaction interfaces
Develop allosteric inhibitors affecting conformational dynamics
C. glabrata is known for its resistance to azole antifungals, often through upregulation of drug efflux pumps . If SNX4 regulates the trafficking of these pumps, inhibiting SNX4 function could potentially restore sensitivity to existing antifungals, providing a novel therapeutic approach to combat resistance.
Translating in vitro findings on C. glabrata SNX4 to in vivo infection models presents several challenges:
Physiological differences between laboratory and host conditions:
In vitro media vs. complex host environments (pH, nutrients, oxygen)
Static cultures vs. dynamic host interactions
Absence of host immune factors in vitro
Temperature and stress conditions that differ from standard laboratory settings
Technical challenges for in vivo assessment:
Limited real-time imaging capabilities in host tissues
Difficulty isolating fungal material from host tissues for molecular analysis
Low fungal burden in some infection models limiting detection sensitivity
Host variability introducing confounding factors
Genetic manipulation considerations:
Stability of genetic modifications in vivo without selection pressure
Potential fitness costs of mutations becoming apparent only in vivo
Different phenotypes in laboratory vs. clinical isolates
Compensatory mechanisms activated specifically in vivo
Experimental design complexities:
Selection of appropriate animal models mimicking human infection
Relevant endpoints for virulence assessment
Controlling for host factors (immune status, microbiome)
Ethical considerations limiting sample sizes and experiment duration
To address these challenges, researchers should consider:
Using ex vivo models as intermediates between in vitro and in vivo
Developing tissue-mimicking culture systems
Employing conditional gene expression systems
Conducting parallel studies in multiple C. glabrata clinical isolates
Novel technologies that could advance our understanding of C. glabrata SNX4 function include:
Advanced imaging approaches:
Super-resolution microscopy to visualize endosomal structures below the diffraction limit
Lattice light-sheet microscopy for 3D imaging with minimal phototoxicity
Single-molecule tracking to follow SNX4 dynamics in living cells
Correlative light and electron microscopy to link function with ultrastructure
Genome editing and screening technologies:
CRISPR interference for tunable gene repression
CRISPR activation for controlled overexpression
Genome-wide CRISPR screens to identify genetic interactions
Inducible degradation systems for acute protein depletion
Protein interaction mapping approaches:
BioID or TurboID proximity labeling in living C. glabrata cells
Thermal proteome profiling to detect drug-target engagement
Cross-linking mass spectrometry to capture transient interactions
Microfluidic techniques for measuring weak interactions
Systems biology approaches:
Multi-omics integration (proteomics, lipidomics, transcriptomics)
Machine learning for pattern recognition in complex datasets
Flux analysis to track membrane protein movement
Computational modeling of trafficking networks
In neurons, a combination of imaging, functional studies, and quantitative proteomics revealed SNX4's unexpected role in synaptic function beyond traditional cargo trafficking . Similar multidisciplinary approaches could uncover the unique aspects of SNX4 function in C. glabrata pathobiology.
Comparative studies across fungal species can significantly inform C. glabrata SNX4 research:
Evolutionary insights:
Sequence conservation analysis to identify functional hotspots
Lineage-specific adaptations in pathogenic vs. non-pathogenic fungi
Rates of evolution in different protein domains suggesting selective pressures
Comparative genomics to identify species-specific interaction partners
Functional conservation testing:
Cross-species complementation studies (e.g., C. glabrata SNX4 in S. cerevisiae snx4Δ)
Chimeric protein construction to map species-specific functional regions
Heterologous expression studies to identify differential localization patterns
Cargo specificity comparison across fungal species
Pathogenesis-specific adaptations:
Comparison between multiple Candida species (C. glabrata, C. albicans, C. krusei)
Analysis of SNX4 function in other pathogenic fungi (Cryptococcus, Aspergillus)
Correlation of SNX4 sequence variations with pathogenicity traits
Host adaptation signatures in SNX4 sequences
Methodological advantages:
Leveraging genetic tools available in model fungi (S. cerevisiae)
Applying insights from well-studied systems to C. glabrata
Identifying conserved cargoes across species
Using phylogenetic relationships to predict function
In S. cerevisiae, SNX4 (Snx4p) prevents degradation of the exocytic v-SNARE Snc1p by recycling it from endosomes to the plasma membrane . Investigating whether C. glabrata SNX4 performs similar functions, and whether additional pathogen-specific cargoes exist, could provide important insights into its role in virulence and stress adaptation.
Critical knowledge gaps in our understanding of C. glabrata SNX4 include:
Basic characterization:
Complete protein structure and domain organization
Subcellular localization pattern in C. glabrata
Expression changes during different growth phases and stress conditions
Post-translational modifications regulating activity
Functional aspects:
Identity of specific cargo proteins in C. glabrata
Composition of SNX4-containing protein complexes
Membrane tubulation and trafficking capabilities
Redundancy with other sorting nexins in C. glabrata
Pathobiology relevance:
Role in virulence and host-pathogen interactions
Contribution to antifungal drug resistance mechanisms
Function during different stages of infection
Impact on stress response and adaptation to host environments
Therapeutic potential:
Essentiality for C. glabrata survival or virulence
Druggability of SNX4 domains or interactions
Potential for combination therapy approaches
Selectivity potential between fungal and human homologs