SPAC11D3.19 is a hypothetical protein encoded by the gene locus SPAC11D3.19, located in chromosome I of S. pombe. While its precise molecular function remains unverified, neighboring genes and conserved domains provide indirect clues:
SPAC11D3.19 resides within a genomic cluster containing genes involved in transport and signaling, suggesting potential roles in cellular homeostasis or stress response.
SPAC11D3.19 is not listed in genome-wide screens for oxidative stress, autophagy, or cytoskeletal regulation , implying it may not be a core component of these pathways.
Codon Optimization: Essential for heterologous expression in E. coli .
Affinity Tagging: His₆ or Smt3 fusion systems to facilitate purification .
SPAC11D3.19 lacks orthologs in Saccharomyces cerevisiae or metazoans, classifying it as a S. pombe-specific protein. This contrasts with conserved Mediator complex subunits (e.g., spMed4/spMed8), which share homology across eukaryotes .
Given its genomic neighborhood and absence from essential gene datasets , SPAC11D3.19 may:
Act as a regulatory accessory in transmembrane signaling.
Participate in non-essential stress adaptation mechanisms.
Subcellular localization.
Interaction partners (e.g., via yeast two-hybrid screens).
Phenotypic consequences of gene deletion.
KEGG: spo:SPAC11D3.19
Initial characterization of SPAC11D3.19 should employ a systematic workflow combining genetic and biochemical techniques. Begin with sequence homology analysis against characterized proteins, followed by gene disruption experiments to observe phenotypic effects.
The recommended methodology for S. pombe genetic manipulation includes:
Design primers to amplify selection markers (such as ura4+ or kanMX) flanked by sequences homologous to regions surrounding SPAC11D3.19
Transform the PCR product into S. pombe cells using standard lithium acetate method
Select transformants on appropriate selective media
Verify gene deletion through PCR using primers binding outside the targeted region
Perform phenotypic analysis under standard and stress conditions
Similar approaches have been successfully employed for characterizing other S. pombe genes. For instance, researchers studying laboratory-strain-specific loss-of-function variants have utilized comparable methodologies for genes like SPAC11D3.11c, which appears to be strain-specific .
For recombinant expression of SPAC11D3.19, a methodical approach incorporating multiple expression systems is recommended:
PCR amplify the SPAC11D3.19 coding sequence from S. pombe genomic DNA
Clone into expression vectors with different affinity tags (His6, GST, MBP) to improve solubility
Test expression in multiple systems:
E. coli BL21(DE3) for bacterial expression
P. pastoris for yeast expression
Baculovirus-insect cell system for eukaryotic post-translational modifications
Optimize expression conditions systematically:
Temperature (16°C, 25°C, 30°C)
Induction time (4h, 8h, overnight)
Inducer concentration
Purify using affinity chromatography followed by size exclusion chromatography
When designing these expression experiments, ensure proper controls are included to validate protein functionality. Include both positive and negative controls to ensure reliability and validity of results .
To predict functional domains of SPAC11D3.19, employ a multi-tiered bioinformatic approach:
Primary sequence analysis:
BLAST against characterized proteins in multiple organisms
PFAM, SMART, and InterPro for conserved domain identification
TMHMM for transmembrane region prediction
SignalP for signal peptide detection
Secondary structure prediction:
PSIPRED for alpha helices and beta sheets
DISOPRED for disordered regions
NetPhos for potential phosphorylation sites
Tertiary structure prediction:
AlphaFold2 for 3D structure modeling
ConSurf for evolutionary conservation mapping onto structure
CASTp for binding pocket prediction
Comparative genomics:
Orthologs identification across fungal species
Synteny analysis to identify conserved genomic context
When analyzing uncharacterized proteins in S. pombe, comparative genomics approaches are particularly valuable. The extensive genomic mapping of S. pombe provides a robust framework for predicting functional relationships .
To determine subcellular localization, implement fluorescent protein tagging with appropriate controls:
Design endogenous tagging construct:
C-terminal GFP/mCherry fusion using PCR-based gene targeting
Include a flexible linker (GGGGS)3 between protein and tag
Maintain the native promoter to preserve physiological expression levels
Create control constructs:
Known nuclear protein (e.g., Gar2-mCherry)
Known ER marker (e.g., Erg11-GFP)
Known Golgi marker (e.g., Anp1-mCherry)
Transformation and verification:
Transform into S. pombe using lithium acetate method
Select transformants on appropriate media
Verify correct integration by PCR and sequencing
Confirm protein functionality through complementation tests
Microscopy and analysis:
Visualize cells at different cell cycle stages
Test multiple growth conditions (standard, stress)
Perform z-stack imaging for 3D localization
Quantify co-localization with organelle markers
This approach provides robust evidence for protein localization while ensuring the tagged protein maintains functionality. When designing these experiments, include appropriate controls for each experimental variable to ensure reliable results .
To identify protein interaction partners, implement complementary approaches:
Affinity Purification coupled with Mass Spectrometry (AP-MS):
Generate S. pombe strain expressing SPAC11D3.19 with tandem affinity tag (TAP)
Perform purification under native conditions
Analyze co-purifying proteins by LC-MS/MS
Filter against control purifications to identify specific interactors
Proximity-dependent Biotin Identification (BioID):
Create SPAC11D3.19-BirA* fusion
Express in S. pombe and induce biotinylation
Purify biotinylated proteins and identify by mass spectrometry
Map interaction network through spatial proximity
Yeast Two-Hybrid screening:
Use SPAC11D3.19 as bait against S. pombe cDNA library
Filter out auto-activators and false positives
Validate interactions through co-immunoprecipitation
Data analysis and validation:
Compare results across different methods
Prioritize proteins identified in multiple approaches
Validate key interactions through reciprocal tagging
Perform functional assays to confirm biological relevance
When analyzing protein interaction data from S. pombe, compare your results with existing datasets to identify patterns and potential functional relationships .
Bulk segregant analysis (BSA) provides a powerful approach for linking phenotypes to genetic variants:
Experimental setup:
Cross SPAC11D3.19 mutant strain with wild-type of opposite mating type
Induce sporulation and isolate random spores
Phenotype progeny and pool based on phenotypic extremes
Extract DNA from pools for whole-genome sequencing
Sequencing and bioinformatic pipeline:
Perform paired-end sequencing (>30x coverage)
Map reads to reference genome using BWA-MEM
Remove duplicate reads using SAMtools' rmdup command
Perform variant calling with SAMtools (options: -B -q 10 -m 3 -F 0.2)
Filter variants based on quality (score ≥30) and read depth (10-200)
Allele frequency analysis:
Calculate reference allele frequencies at each SNP position
Generate scatter plots of allele frequencies between pools
Apply LOESS regression to visualize trends (span parameter ~60kb)
Identify genomic regions with skewed segregation
This methodology has been validated for uncovering trait-gene relationships in fission yeast strains . When implementing BSA, careful experimental design and rigorous statistical analysis are essential to identify significant genetic associations.
If bioinformatic analysis suggests DNA-binding properties for SPAC11D3.19, implement ChIP-seq with these methodological considerations:
Experimental design:
Generate strain with epitope-tagged SPAC11D3.19 (HA, FLAG)
Include untagged control and input samples
Prepare minimum 2 biological replicates
Test multiple growth conditions if appropriate
ChIP protocol optimization:
Crosslink cells with 1% formaldehyde for 15 minutes
Lyse cells and sonicate to 200-500bp fragments
Immunoprecipitate using antibodies against tag
Include mock IP controls
Reverse crosslinks and purify DNA
Sequencing and analysis pipeline:
Prepare libraries and sequence (minimum 20M reads)
Map reads to S. pombe genome
Call peaks with at least 2-fold enrichment over input
Identify enriched motifs using MEME-ChIP
Correlate binding sites with gene expression data
Based on ChIP-seq studies in S. pombe, most transcription factor binding sites reside in accessible regions with low histone H3 levels and elevated H3K14ac . The number of binding sites can vary widely between different DNA-binding proteins, ranging from 1 to 356 sites per factor .
For precise genome editing of SPAC11D3.19, implement CRISPR-Cas9 with these methodological considerations:
Guide RNA design:
Identify target sites using S. pombe-specific CRISPR design tools
Select guides with minimal off-target effects
Design multiple guides targeting different regions
Include positive control guides targeting known sites
Repair template design:
For point mutations: 60-80bp homology arms surrounding the mutation
For insertions/tags: longer homology arms (>500bp)
Include silent mutations in PAM site to prevent re-cutting
Transformation and screening:
Co-transform Cas9, gRNA, and repair template
Use drug selection markers when possible
Screen transformants by PCR, restriction digest, or sequencing
Verify off-target effects in top predicted sites
Validation of edited strains:
Sequence the entire target locus
Confirm expression levels are maintained
Verify protein function through complementation tests
Test phenotypes under multiple conditions
This approach allows for precise modifications including point mutations, deletions, insertions, or tagging without introducing selection markers that might affect adjacent genes.
When studying phenotypic effects, implement these essential controls:
Strain controls:
Wild-type parental strain
Independent deletion clones (minimum 3)
Complementation strain (deletion with reintroduced functional gene)
Marker-only control (selection marker at neutral locus)
Experimental controls:
Positive control (strain with known phenotype)
Negative control (strain without phenotype)
Technical replicates for quantitative measurements
Biological replicates (minimum 3 independent experiments)
Phenotypic analysis matrix:
Standard conditions (YES, EMM media)
Stress conditions (temperature, pH, oxidative, osmotic)
Cell cycle analysis (synchronization by nitrogen starvation)
Growth rate determination (growth curves, spot assays)
Statistical analysis:
Determine appropriate statistical tests based on data distribution
Calculate p-values with multiple testing correction
Report effect sizes alongside statistical significance
Use power analysis to determine sample size
This comprehensive control strategy ensures that observed phenotypes are specifically attributable to SPAC11D3.19 deletion rather than background effects or experimental artifacts .
For robust analysis of differential expression data:
Pre-processing and quality control:
Filter low-count genes (minimum 10 reads in at least 3 samples)
Assess sample-to-sample variation with PCA
Identify and handle batch effects
Normalize counts appropriately (DESeq2, TMM, or quantile normalization)
Statistical testing framework:
For RNA-seq: negative binomial models (DESeq2, edgeR)
For proteomics: linear models with empirical Bayes (limma)
Multiple testing correction (Benjamini-Hochberg FDR)
Significance thresholds (padj < 0.05, |log2FC| > 1)
Advanced analytical approaches:
Gene set enrichment analysis (GSEA)
Pathway analysis (ReactomeFI, STRING)
Co-expression network analysis (WGCNA)
Integration with ChIP-seq or proteomics data
Visualization and reporting:
MA plots for global expression changes
Volcano plots for significance vs. magnitude
Heatmaps for gene clusters
Pathway diagrams for functional context
When working with S. pombe expression data, data.table provides efficient methods for manipulating large datasets . For example:
This approach allows for efficient computation across specific gene columns grouped by experimental conditions .
To integrate multi-omics data for comprehensive functional characterization:
Data collection and standardization:
Ensure comparable experimental conditions across platforms
Transform data to comparable scales
Account for different dynamic ranges between technologies
Address missing values appropriately
Correlation analysis:
Calculate protein-mRNA correlation coefficients
Identify discordant expression patterns
Cluster genes/proteins with similar behavior
Test for post-transcriptional regulation
Pathway and network integration:
Map data onto known pathways
Construct integrated networks
Identify functional modules
Predict regulatory relationships
Visualization and interpretation:
Multi-layered network visualization
Integrated heatmaps
Biological context mapping
Hypothesis generation
When integrating data for S. pombe proteins, context from existing datasets is valuable. For instance, compare your findings with the list of genes showing significant transcriptional changes under relevant conditions, as illustrated in Table 7 from genomic studies in S. pombe :
Gene ID | Gene Symbol | Gene Function |
---|---|---|
SPAC212.11 | tlh1 | RecQ type DNA helicase |
SPAC19G12.16c | adg2 | conserved fungal cell surface protein |
SPBC1348.14c | ght7 | plasma membrane hexose transmembrane transporter |
SPAPB1E7.04c | SPAPB1E7.04c | chitinase |
SPBC1105.05 | exg1 | cell wall glucan 1,6-beta-glucosidase |
SPAC1039.11c | gto1 | alpha-glucosidase |
SPAC186.09 | pdc102 | pyruvate decarboxylase |
SPAC19B12.02c | gas1 | cell wall 1,3-beta-glucanosyltransferase |
SPBC4F6.12 | pxl1 | paxillin-like protein |
SPAC1F8.05 | isp3 | spore wall structural constituent |
SPAC750.01 | SPAC750.01 | NADP-dependent aldo/keto reductase |
To investigate potential protein complex involvement:
Size exclusion chromatography with western blotting:
Prepare native cell extracts from tagged SPAC11D3.19 strain
Fractionate by size exclusion chromatography
Analyze fractions by western blotting
Compare elution profile with known complex markers
Co-immunoprecipitation with mass spectrometry:
Optimize lysis conditions to maintain complex integrity
Perform IP with antibodies against tagged SPAC11D3.19
Identify co-purifying proteins by mass spectrometry
Compare with control IPs to identify specific interactors
Blue native PAGE analysis:
Prepare native complexes from tagged strains
Separate on blue native gels
Identify complex components by mass spectrometry
Perform supershift assays with specific antibodies
Density gradient ultracentrifugation:
Prepare cell extracts under native conditions
Separate complexes by sucrose or glycerol gradient
Analyze fractions for SPAC11D3.19 and potential complex members
Compare with known complex markers
When analyzing potential protein complexes, compare elution profiles or interaction partners with known S. pombe complexes to identify functional relationships .
When publishing research on uncharacterized proteins like SPAC11D3.19, follow these best practices:
Comprehensive characterization approach:
Combine multiple methodologies (genetic, biochemical, -omics)
Provide both in vivo and in vitro evidence
Include thorough controls and statistical analysis
Address potential functional redundancy
Data sharing and reproducibility:
Deposit raw data in appropriate repositories (GEO, ProteomeXchange)
Provide detailed protocols with specific reagent information
Make strains available through repositories (Yeast Genetic Resource Center)
Include sufficient methodological detail for reproduction
Nomenclature and annotation:
Follow community standards for gene/protein naming
Provide evidence levels for functional assignments
Update database entries with new findings
Clearly distinguish between direct observations and predictions
Contextual interpretation:
Relate findings to known biological processes
Discuss evolutionary conservation
Compare with related proteins in S. pombe
Connect to broader biological significance