Recombinant YKL165C-A is synthesized using optimized expression vectors and purification protocols:
Expression Vectors:
Purification:
Host System | Advantages | Limitations |
---|---|---|
E. coli | High yield, low cost | Limited post-translational modifications |
Yeast | Eukaryotic protein folding, glycosylation | Lower yield compared to E. coli |
Mammalian Cells | Native-like post-translational modifications | High cost, complex protocols |
ELISA Kits: Commercially available for detecting YKL165C-A-specific antibodies .
Western Blot: Used to validate protein expression in recombinant yeast strains .
YKL165C-A remains poorly characterized in functional studies. Notably:
Genomic Context: Localized to chromosome IV in S. cerevisiae .
Orthologs: No closely related homologs identified in non-fungal species .
Hypothesized Roles:
STRING: 4932.YKL165C-A
YKL165C-A is a putative uncharacterized protein from Saccharomyces cerevisiae (baker's yeast). It consists of 77 amino acids with the following sequence: MQFPVFFFRCFSYGISSMPLKNKVVFNENMERKDTFYQLILKVLSALLLLSVRNSSGHTR HFVQSSEKIYRRSLFKQ . The protein is classified as "putative uncharacterized," indicating that its precise function has not been definitively established. Current structural data is limited, with no published crystal structure available. Researchers typically work with recombinant forms of the protein, often expressed in E. coli with affinity tags (such as His-tag) to facilitate purification and subsequent analysis .
Recombinant YKL165C-A can be obtained through several approaches:
Commercial sources: Pre-made recombinant protein is available from specialized suppliers who provide His-tagged full-length YKL165C-A (1-77 amino acids) expressed in E. coli systems .
In-house expression: Researchers can generate their own recombinant YKL165C-A by:
Designing and synthesizing or acquiring the gene sequence
Cloning into an appropriate expression vector with desired fusion tags
Transforming into a suitable expression system (typically E. coli)
Inducing expression under optimized conditions
Purifying using affinity chromatography based on the fusion tag
For reproducing experimental results, it is important to document the exact source and preparation method of the protein, as variations in expression systems can affect post-translational modifications and protein folding.
While E. coli is the most commonly used expression system for recombinant YKL165C-A , researchers should consider the following systems based on experimental requirements:
Expression System | Advantages | Limitations | Recommended for |
---|---|---|---|
E. coli | High yield, cost-effective, rapid expression | Limited post-translational modifications, potential inclusion body formation | Structural studies, antibody production, initial characterization |
Native S. cerevisiae | Natural post-translational modifications, proper folding | Lower yields, more complex purification | Functional studies, protein-protein interaction analysis |
Pichia pastoris | Higher eukaryotic modifications, secretion possible | Longer development time | Studies requiring proper glycosylation |
Cell-free systems | Rapid production, avoids toxicity issues | Higher cost, smaller scale | Quick screening experiments |
The choice of expression system should be guided by research objectives. For studying YKL165C-A function in its native context, expression in S. cerevisiae with minimal tags is often preferable despite potentially lower yields.
Characterizing uncharacterized proteins like YKL165C-A requires a systematic experimental approach. Begin by defining clear research questions and variables related to potential functions . A recommended experimental workflow includes:
Sequence analysis and prediction:
Conduct bioinformatic analysis for conserved domains
Perform phylogenetic comparisons with characterized proteins
Use structure prediction algorithms to inform functional hypotheses
Localization studies:
Interaction studies:
Implement yeast two-hybrid screens to identify potential binding partners
Confirm interactions using co-immunoprecipitation and pull-down assays
Create interaction networks to situate the protein in cellular pathways
Loss-of-function studies:
Generate knockout strains using CRISPR or traditional deletion techniques
Perform comprehensive phenotypic characterization under various conditions
Implement rescue experiments with wild-type and mutant constructs
Each step should include appropriate controls and replications to ensure reliable results. Documenting negative results is equally important, as they can provide valuable insights into what the protein does not do 6.
When investigating protein-protein interactions involving YKL165C-A, multiple complementary approaches should be employed:
In vivo techniques:
Yeast two-hybrid (Y2H) screening using YKL165C-A as bait against a yeast library
Proximity-dependent biotin identification (BioID) to identify neighboring proteins
Fluorescence resonance energy transfer (FRET) to confirm direct interactions in live cells
In vitro techniques:
Validation strategies:
Co-immunoprecipitation from native yeast cells
Reciprocal tagging of potential interactors
Competition assays with purified domains
Each method has distinct advantages and limitations. For example, Y2H may detect transient interactions but can generate false positives, while co-IP confirms interactions in native contexts but may miss weak or transient associations. Therefore, using multiple methods strengthens confidence in identified interactions. Document all experimental conditions thoroughly, including buffer compositions, temperature, and protein concentrations, as these can significantly affect interaction detection6.
Designing controlled experiments to study YKL165C-A knockouts requires careful consideration of variables and appropriate controls . A systematic approach would include:
Strain development:
Generate YKL165C-A deletion strain using homologous recombination or CRISPR-Cas9
Create complemented strains reintroducing wild-type YKL165C-A
Develop site-directed mutants to test specific functional hypotheses
Ensure genetic background is consistent across all strains
Experimental design considerations:
Use between-subjects design comparing wild-type, knockout, and complemented strains
Control extraneous variables such as media composition, temperature, and growth phase
Include technical replicates (minimum 3) and biological replicates (minimum 3)
Implement blind analysis when possible to reduce experimenter bias 6
Phenotypic characterization matrix:
Condition | Parameters to Measure | Methods | Data Analysis |
---|---|---|---|
Standard growth | Growth rate, cell morphology | Growth curves, microscopy | ANOVA, growth curve fitting |
Stress conditions (temperature, pH, osmotic) | Survival rate, stress response genes | Spot assays, qRT-PCR | Survival analysis, expression fold change |
Metabolic challenges | Metabolite utilization, enzyme activities | Metabolomics, enzymatic assays | Pathway analysis, PCA |
Cell cycle perturbations | Cell cycle progression, DNA content | Flow cytometry, synchronization studies | Cell cycle distribution analysis |
Validation approaches:
Rescue experiments reintroducing YKL165C-A under native or inducible promoters
Cross-complementation with homologs from related species
Dose-response relationships with controlled expression levels
When reporting results, clearly distinguish between direct phenotypic effects and potential secondary effects due to compensatory mechanisms or strain adaptation .
Elucidating YKL165C-A function requires sophisticated proteomic approaches that can provide insights into its dynamic behavior and interactome:
Proximity-based labeling techniques:
BioID fusion to YKL165C-A to identify proximal proteins through biotinylation
APEX2 labeling for temporal resolution of interaction dynamics
Split-BioID for conditional proximity mapping
Quantitative interaction proteomics:
SILAC (Stable Isotope Labeling with Amino acids in Cell culture) combined with IP-MS
TMT (Tandem Mass Tag) labeling for multiplexed comparison across conditions
Label-free quantification for native protein complexes
Structural proteomics:
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify dynamic regions
Cross-linking mass spectrometry (XL-MS) to map protein interfaces
Native mass spectrometry to analyze intact complexes
Global proteome impact assessment:
Comparative proteomics between wild-type and YKL165C-A deletion strains
Phosphoproteomics to detect signaling pathway alterations
Thermal proteome profiling to identify proteins affected by YKL165C-A deletion
For each approach, establish rigorous statistical frameworks for data analysis, including appropriate normalization methods, significance thresholds, and multiple testing corrections. Cross-validate findings using orthogonal techniques and integrate datasets to build comprehensive functional models. Consider temporal dynamics by sampling at multiple timepoints, particularly following perturbations that might engage YKL165C-A-dependent pathways.
Resolving contradictory data about YKL165C-A requires a systematic approach to identify sources of variability and reconcile disparate findings:
Methodological reconciliation:
Compare experimental conditions in detail (strain backgrounds, growth media, temperature)
Evaluate tag positions and types (N-terminal vs. C-terminal, size of tags)
Assess detection methods (direct fluorescence vs. immunofluorescence, antibody specificity)
Consider temporal aspects (cell cycle stage, growth phase, induction conditions)
Technical validation strategies:
Implement orthogonal techniques to confirm findings (e.g., both microscopy and biochemical fractionation for localization)
Vary tag orientation and type to rule out tag interference
Use complementary antibodies targeting different epitopes
Perform rescue experiments with untagged constructs
Biological context analysis:
Test for condition-specific behaviors (stress conditions, nutrient availability)
Investigate cell-to-cell variability through single-cell approaches
Examine potential moonlighting functions in different cellular compartments
Consider post-translational modifications that might affect localization or function
Collaborative resolution approaches:
Arrange direct comparison experiments between laboratories reporting contradictory results
Exchange key reagents (strains, antibodies, constructs) to rule out reagent variability
Standardize protocols and establish minimal reporting requirements
Computational approaches provide valuable insights for predicting YKL165C-A function and strategically guiding experimental design:
Sequence-based predictions:
Profile Hidden Markov Models to identify remote homology
Conservation analysis across yeast species to identify functional constraints
Coevolution analysis to predict interaction interfaces
Disorder prediction to identify flexible regions potentially involved in interactions
Structural modeling:
Ab initio modeling for domains lacking homology to known structures
Molecular dynamics simulations to predict conformational flexibility
Binding site prediction to identify potential ligand pockets
Protein-protein docking with predicted interaction partners
Systems-level analyses:
Gene neighborhood analysis and synteny across species
Coexpression network analysis to identify functionally related genes
Genetic interaction profiles comparison with genes of known function
Metabolic modeling to predict involvement in specific pathways
Machine learning applications:
Functional prediction using ensemble methods integrating multiple features
Transfer learning from characterized proteins to YKL165C-A
Deep learning on structural features to predict binding sites
A strategic implementation would involve:
Generating multiple computational hypotheses
Ranking predictions by confidence scores
Designing targeted experiments to test specific computational predictions
Iteratively refining models based on experimental feedback
Researchers working with YKL165C-A frequently encounter several technical challenges that can be addressed through methodological refinements:
Protein solubility and stability issues:
Challenge: Recombinant YKL165C-A may form aggregates or inclusion bodies during expression
Solutions:
Optimize expression temperature (typically lowering to 16-18°C)
Test multiple solubility tags (MBP, SUMO, Thioredoxin)
Incorporate stabilizing additives in buffers (glycerol, arginine, low concentrations of detergents)
Consider native purification from S. cerevisiae despite lower yields
Antibody specificity problems:
Challenge: Generating specific antibodies against small proteins like YKL165C-A (77 amino acids)
Solutions:
Use epitope mapping to identify unique regions
Validate antibodies using knockout strains as negative controls
Consider alternative detection methods (e.g., epitope tagging)
Implement sandwich ELISA approaches for improved specificity
Expression level detection:
Challenge: Low endogenous expression levels making detection difficult
Solutions:
Employ sensitive detection methods (nested PCR, digital PCR for mRNA)
Use signal amplification methods for protein detection
Consider concentration steps before analysis
Optimize induction conditions for recombinant expression
Functional redundancy masking phenotypes:
Challenge: Lack of clear phenotypes in single gene deletions due to compensatory mechanisms
Solutions:
Create combinatorial knockouts with related genes
Test phenotypes under diverse stress conditions
Use sensitized genetic backgrounds
Implement acute depletion systems (e.g., degron tags) to bypass compensation
For each challenge, systematic optimization should be documented thoroughly to establish reproducible protocols. Collaborative approaches, sharing both successful and failed strategies across research groups, can accelerate technical problem-solving in this field.
Optimizing experimental conditions for studying YKL165C-A protein-protein interactions requires systematic parameter adjustment and validation:
Buffer optimization strategy:
Test multiple buffer compositions systematically:
Buffer Component | Range to Test | Considerations |
---|---|---|
pH | 6.0-8.0 in 0.5 increments | Match physiological pH of compartment where YKL165C-A localizes |
Salt (NaCl) | 50-300 mM | Higher concentrations reduce non-specific interactions but may disrupt weak specific interactions |
Detergents | 0.01-0.1% non-ionic detergents | Critical for membrane-associated interactions but can disrupt some complexes |
Stabilizers | 5-10% glycerol, 1-5 mM TCEP | Maintain protein stability during experiments |
Divalent cations | 1-5 mM MgCl₂, CaCl₂ | May be required for specific interactions |
Protein state considerations:
Compare native extraction vs. recombinant proteins
Test tag position effects (N-terminal vs. C-terminal)
Evaluate effect of post-translational modifications on interactions
Control protein concentration ratios in binding assays
Interaction detection optimization:
Adjust incubation times (15 min to overnight) and temperatures (4°C to 30°C)
Optimize washing stringency in pull-down assays
Compare different detection methods (Western blot vs. mass spectrometry)
Implement crosslinking strategies for transient interactions
Validation controls:
Include known interacting protein pairs as positive controls
Use unrelated proteins with similar biochemical properties as negative controls
Perform competition assays with unlabeled proteins
Test interaction dependency on specific domains through truncation constructs
For each interaction identified, establish a confidence score based on reproducibility across methods, detection under multiple conditions, and validation through orthogonal approaches. Record all optimization steps in detail to ensure reproducibility and to inform future interaction studies with YKL165C-A.
Several cutting-edge technologies are poised to significantly advance our understanding of YKL165C-A function:
Advanced imaging technologies:
Super-resolution microscopy (PALM/STORM) for precise localization studies
Cryo-electron tomography to visualize YKL165C-A in its native cellular context
Live-cell single-molecule tracking to monitor dynamics and interactions
Correlative light and electron microscopy (CLEM) to bridge molecular and ultrastructural information
Genome engineering advances:
CRISPR base editing for precise point mutations without DNA breaks
CRISPRi/CRISPRa for tunable expression modulation
Genome-wide interaction screens using CRISPR-based approaches
Site-specific recombination systems for controlled temporal studies
Single-cell technologies:
Single-cell proteomics to examine YKL165C-A expression heterogeneity
Single-cell metabolomics to link YKL165C-A to metabolic phenotypes
Multi-omics integration at single-cell resolution
Microfluidic systems for temporally resolved single-cell studies
Structural biology innovations:
Cryo-EM for structural determination of YKL165C-A complexes
Integrative structural biology combining multiple data types
AlphaFold2 and related AI approaches for structure prediction
Time-resolved structural methods to capture conformational changes
To effectively leverage these technologies, researchers should focus on collaborative approaches that combine technical expertise across disciplines. Consider establishing consortia or collaborative networks specifically focused on uncharacterized yeast proteins like YKL165C-A, pooling resources and standardizing protocols to accelerate discoveries.
Systems biology approaches provide powerful frameworks for understanding YKL165C-A within the broader context of cellular networks:
Network integration strategies:
Construct multi-layered networks incorporating:
Protein-protein interaction data
Genetic interaction profiles
Transcriptional regulation networks
Metabolic pathway connections
Apply network analysis algorithms to identify:
Network motifs involving YKL165C-A
Centrality measures to assess functional importance
Modularity to identify functional clusters
Perturbation-based approaches:
Implement systematic environmental perturbations:
Chemical genetic profiling under diverse conditions
Temporal response to stress conditions
Nutrient limitation studies
Analyze differential network states:
Condition-specific rewiring involving YKL165C-A
Dynamic changes in interaction partners
Compensatory network adaptations in YKL165C-A mutants
Predictive modeling frameworks:
Develop kinetic models of pathways potentially involving YKL165C-A
Implement constraint-based models to predict metabolic impacts
Apply Bayesian networks to infer causal relationships
Utilize machine learning to predict emergent phenotypes
Multi-omics data integration:
Correlate protein abundance, localization, modification, and interactions
Identify condition-specific activation of YKL165C-A functions
Map impacts of YKL165C-A perturbation across biological scales
Develop visualization tools for multi-dimensional data exploration
The systems biology approach is particularly valuable for uncharacterized proteins like YKL165C-A, as it can reveal functional context even when direct biochemical functions remain unclear. This holistic perspective can illuminate roles in cellular homeostasis, stress responses, or metabolic regulation that might be missed by reductionist approaches alone.