KEGG: dha:DEHA2E18964g
D. hansenii is a halophilic non-conventional yeast found in natural environments with high salt concentrations and osmotic pressure (e.g., seawater, soil, glaciers, and salty food). Its significance for PCC1 research stems from its unique ability to withstand high osmotic pressure, high salinity, and low water activity . As an osmotolerant yeast that can grow in the presence of high concentrations of salt or ethanol, it offers a distinctive cellular environment for studying polarized growth and chromatin-associated proteins like PCC1 .
For initiating D. hansenii cultures, the following methodological approach is recommended:
Media selection: YPD medium (1% yeast extract, 2% peptone, 2% glucose) or YM DEB medium are suitable for routine cultivation .
Growth conditions: Optimal growth occurs at temperatures below 30°C (23-28°C is ideal) .
Adaptation procedure:
Start with standard YM DEB medium
Gradually adapt cells to experimental conditions through sequential subculturing
For starvation experiments, follow the adaptation process shown below:
Adaptation Process for D. hansenii Cultures:
| Stage | Medium | Duration | Purpose |
|---|---|---|---|
| Initial growth | YM DEB | 24-48 hours | Establish viable culture |
| Intermediate adaptation | 50% YM DEB + 50% experimental medium | 24 hours | Gradual adaptation |
| Final adaptation | 100% experimental medium | 24 hours | Complete adaptation |
| Experimental setup | Starvation medium (YM1) | Experimental timeline | Research conditions |
This adaptation process is particularly important when studying stress responses or preparing cells for recombinant protein expression .
When designing primers for PCC1 amplification in D. hansenii, researchers should consider:
Codon usage bias: D. hansenii belongs to the CTG clade, which encodes serine instead of leucine in CTG codons . This necessitates codon optimization of heterologous genes.
Primer design parameters:
Special considerations for PCC1:
Using this approach, targeted integration efficiency of >75% can be achieved through homologous recombination .
The selection of appropriate markers is crucial for successful recombinant protein expression. Based on current research, several effective markers for D. hansenii have been established:
Auxotrophic markers:
Heterologous dominant selectable markers (applicable to wild-type isolates):
Selection marker efficiency comparison:
For PCC1 expression, heterologous markers are generally recommended as they allow work with wild-type D. hansenii strains that may exhibit better growth and stress tolerance characteristics .
Despite previous reports of inefficient homologous recombination in D. hansenii, recent studies demonstrate that high efficiency can be achieved with these methodological improvements:
Flank length optimization:
Selection marker design:
Transformation protocol optimization:
Safe harbor site targeting:
This optimized approach enables efficient targeted genomic modification through homologous recombination in D. hansenii isolates .
Selection of appropriate promoter and terminator sequences significantly impacts recombinant protein expression levels. The following have proven effective in D. hansenii:
Promoters:
Heterologous TEF1 promoter from Arxula adeninivorans - highest production of reporter proteins
Meyerozyma guilliermondii ACT1 promoter (MgACT1pr) - demonstrated to direct GFP expression in D. hansenii
Saccharomyces stipitis GPD1 promoter (500 bp upstream of GPD1 ORF)
D. hansenii TEF1 promoter (DhTEF) - successful for reporter gene expression
Terminators:
For optimal expression, in vivo DNA assembly techniques can be used to screen potential promoters and terminators to enhance D. hansenii's production of recombinant proteins in specific growth conditions .
CRISPR/Cas9 technology has recently been adapted for D. hansenii, providing powerful options for precise gene editing. Key methodological considerations include:
CRISPR system optimization:
PAM site selection:
Target PAM sites that avoid CTG codons to prevent off-target effects
Consider the chromatin structure around the PCC1 locus for accessibility
Repair template design:
Delivery method:
Ribonucleoprotein (RNP) complex delivery may improve efficiency
Plasmid-based expression systems using ARS elements for transient Cas9 expression
Strain selection:
The CRISPR/Cas9 method allows for precise gene editing, which is essential for the development of D. hansenii as a cell factory for various biotechnological applications, including controlled expression of proteins like PCC1 .
D. hansenii exhibits significant genetic heterogeneity and potential ploidy variations that complicate genetic studies. To address these challenges:
Strain characterization:
Multi-strain approach:
Confirmation of gene disruption:
Strain-specific optimization:
Adjust transformation and selection protocols for each strain
Consider that homologous recombination efficiency varies between strains
Genetic heterogeneity analysis example:
| D. hansenii Strain | Chromosome Pattern | Genetic Similarity | Phenotypic Variation | Recommended Approach |
|---|---|---|---|---|
| CBS 767 (reference) | Standard karyotype | Reference | Moderate salt tolerance | Standard protocols |
| Strain A | Variant karyotype | 85% similarity to reference | High salt tolerance | Increased homology arms |
| Strain B | Variant karyotype | 70% similarity to reference | Low salt tolerance | Alternative selection markers |
This approach accounts for the significant genetic heterogeneity among D. hansenii strains, which can be reflected in intra-species variation at the phenotypic level .
Understanding the subcellular localization and dynamics of PCC1 requires sophisticated imaging techniques. Recent advances applicable to D. hansenii include:
High-resolution live cell fluorescent imaging:
Label-free holotomography:
Combined approaches:
Integrate fluorescent imaging with holotomography for comprehensive analysis
Correlative light and electron microscopy (CLEM) to combine fluorescence imaging with ultrastructural details
Protein detection methods:
This combinatorial approach serves as a template for studying cell biological systems like D. hansenii that are not amenable to standard genetic procedures .
To effectively study PCC1's role in osmotic stress response, a comprehensive experimental design approach is recommended:
Strain construction:
Stress conditions setup:
Multi-omics analysis:
Transcriptomic analysis to identify PCC1-dependent gene expression changes
Proteomic and phosphoproteomic analyses to detect post-translational modifications
Metabolomic profiling to understand metabolic adaptations
Experimental matrix design:
| Strain Type | Control Condition | Stress Condition 1 | Stress Condition 2 | Stress Condition 3 |
|---|---|---|---|---|
| Wild-type | No salt | 1M NaCl | 1M KCl | 2M Sorbitol |
| PCC1Δ | No salt | 1M NaCl | 1M KCl | 2M Sorbitol |
| PCC1-GFP | No salt | 1M NaCl | 1M KCl | 2M Sorbitol |
| Conditional PCC1 | No salt, ±inducer | 1M NaCl, ±inducer | 1M KCl, ±inducer | 2M Sorbitol, ±inducer |
Key parameters to measure:
This experimental design will provide comprehensive insights into PCC1's role in D. hansenii's remarkable osmotolerance and stress adaptation capabilities .
To effectively study PCC1's interaction with chromatin in D. hansenii, researchers should implement this methodological framework:
Chromatin immunoprecipitation (ChIP) protocol optimization:
Generate epitope-tagged PCC1 strains (e.g., PCC1-FLAG, PCC1-HA)
Optimize cell fixation conditions for D. hansenii's robust cell wall
Develop chromatin fragmentation protocols specific to D. hansenii's chromatin structure
Validate antibody specificity and efficiency in D. hansenii context
Chromatin interaction mapping:
Perform ChIP-seq to identify genome-wide binding sites of PCC1
Implement ChIP-exo or CUT&RUN for higher resolution binding site mapping
Correlate binding sites with gene expression data to identify direct regulatory targets
Protein-protein interaction analysis:
Perform co-immunoprecipitation to identify PCC1 interaction partners
Validate interactions through reciprocal pull-downs
Use proximity labeling approaches (BioID, APEX) to identify chromatin-associated interaction partners
Experimental conditions matrix:
| Growth Phase | Standard Condition | Salt Stress | Nutrient Limitation | Temperature Stress |
|---|---|---|---|---|
| Log phase | 28°C, YPD | 1M NaCl | Carbon limitation | 37°C |
| Stationary phase | 28°C, YPD | 1M NaCl | Nitrogen limitation | 4°C |
| Post-stress recovery | After 2h stress exposure | After 2h salt exposure | After 2h starvation | After 2h temperature exposure |
Controls and validations:
Include non-tagged strains as negative controls
Use known chromatin-associated proteins as positive controls
Validate binding sites through reporter assays or mutagenesis
This comprehensive approach will provide insights into how PCC1 interacts with chromatin under different physiological conditions and contributes to D. hansenii's unique stress adaptation capabilities.
High-throughput screening approaches can efficiently identify conditions affecting PCC1 function. An optimal methodological framework includes:
Reporter system development:
Create PCC1-fluorescent protein fusions to monitor expression and localization
Develop promoter-reporter constructs for PCC1-regulated genes
Establish growth-based readouts for PCC1 functionality
Micro-cultivation screening setup:
Experimental condition matrix:
Data analysis and integration:
Validation in controlled bioreactors:
Confirm high-throughput findings in controlled chemostat cultivations
Perform transcriptomic and proteomic analyses of validated conditions
Examine PCC1 localization and activity under validated conditions
Example screening matrix:
| Parameter | Level 1 | Level 2 | Level 3 | Level 4 | Level 5 |
|---|---|---|---|---|---|
| NaCl (M) | 0 | 0.5 | 1.0 | 1.5 | 2.0 |
| pH | 4 | 5 | 6 | 7 | 8 |
| Carbon source | Glucose | Xylose | Glycerol | Sorbitol | Galactose |
| Temperature (°C) | 15 | 20 | 25 | 30 | 37 |
This high-throughput approach allows for systematic identification of conditions that modulate PCC1 function while efficiently managing experimental resources .
When facing low expression of recombinant PCC1, implement this systematic troubleshooting approach:
Expression construct optimization:
Integration site assessment:
Host strain selection:
Evaluate expression in multiple D. hansenii strains due to genetic heterogeneity
Consider using strains with characterized proteolytic profiles
Test expression in protease-deficient strains if available
Culture condition optimization:
Adjust cultivation temperature (optimal growth below 30°C)
Optimize media composition and pH
Test expression in the presence of varying salt concentrations
Monitor growth phase-dependent expression
Protein stability assessment:
Check for proteolytic degradation of expressed PCC1
Consider fusion tags to enhance stability
Implement pulse-chase experiments to determine protein half-life
Systematic troubleshooting matrix:
| Factor | Test 1 | Test 2 | Test 3 | Expected Outcome |
|---|---|---|---|---|
| Promoter | MgACT1pr | TEF1 | DhGPD1 | Identify optimal promoter |
| Integration site | Native locus | DhARG1 | Random | Determine best expression site |
| Host strain | CBS 767 | NCYC102 | NCYC3363 | Identify optimal strain |
| Growth temperature | 23°C | 25°C | 28°C | Optimize expression conditions |
| Salt concentration | 0M | 0.5M | 1M | Determine effect of salt on expression |
This systematic approach will help identify and address the specific factors limiting PCC1 expression in D. hansenii .
When facing contradictory experimental results in PCC1 research, implement this methodological framework to resolve discrepancies:
Genetic heterogeneity assessment:
Experimental condition standardization:
Document precise cultivation conditions (media composition, pH, temperature)
Standardize growth phase for sampling (e.g., mid-log phase at specific OD)
Control environmental factors like salt concentration and osmotic pressure
Implement chemostat cultivation for precise physiological control
Methodological validation:
Perform cross-laboratory validation of key protocols
Include multiple technical and biological replicates
Use complementary methods to confirm findings
Implement appropriate statistical analyses for result validation
Strain-specific effects investigation:
Test PCC1 function across multiple D. hansenii strains
Create isogenic strains differing only in PCC1 alleles
Perform complementation tests with different PCC1 alleles
Systematic literature review and meta-analysis:
Resolution framework matrix:
| Contradiction Type | Primary Investigation | Secondary Investigation | Validation Approach |
|---|---|---|---|
| Growth phenotype | Strain verification, Media standardization | Growth curve in controlled bioreactor | Cross-strain testing |
| Gene expression | RNA extraction method validation | RT-qPCR with multiple reference genes | RNA-seq validation |
| Protein localization | Fixation protocol optimization | Live cell imaging vs. fixed cells | Subcellular fractionation |
| Stress response | Precise stress application timing | Time-course sampling | Metabolite profiling |
This systematic approach acknowledges the significant strain variation in D. hansenii and implements rigorous controls to resolve contradictory results .