YDR182W-A is a hypothetical protein encoded by the YDR182W-A gene in Saccharomyces cerevisiae strain S288C. Hypothetical proteins are computationally predicted but lack experimental validation, often due to insufficient functional data. Key genetic attributes include:
The YDR182W-A gene was identified through fungal homology and RT-PCR analysis, with no known functional annotations in Gene Ontology (GO) databases .
Recombinant YDR182W-A is synthesized in E. coli for research purposes. Key production specifications include:
The protein is marketed for molecular studies, including interaction assays and structural analysis .
YDR182W-A exhibits genetic interactions with critical yeast proteins, though its exact role remains unclear:
Interactor | Interaction Type | SGA Score | Phenotype | Reference |
---|---|---|---|---|
CDC11 | Negative Genetic | -0.1795 | Enhanced fitness defect | BioGRID |
RPB7 | Negative Genetic | -0.1956 | Colony size reduction | BioGRID |
CDC11: A septin ring component essential for cytokinesis; mutations in CDC11 and YDR182W-A synergistically impair cell viability .
RPB7: A subunit of RNA polymerase II; genetic ablation of both genes exacerbates transcriptional defects .
These interactions suggest YDR182W-A may indirectly influence cellular processes like cytokinesis or transcriptional regulation.
Despite its recombinant availability, YDR182W-A lacks experimental validation for:
Expression Patterns: No microarray or RNA-seq data exist for YDR182W-A in yeast .
Subcellular Localization: No evidence of organelle-specific targeting or membrane association.
Biochemical Activity: No enzymatic function (e.g., phosphatase, kinase) has been reported .
Conservation: Limited homology to characterized proteins in other fungi or eukaryotes .
The recombinant protein is used primarily for:
Interaction Studies: Co-IP or affinity chromatography to identify binding partners .
Structural Analysis: X-ray crystallography or NMR to determine 3D conformation .
Control Experiments: Negative control in assays requiring uncharacterized proteins .
KEGG: sce:YDR182W-A
STRING: 4932.YDR182W-A
YDR182W-A is an uncharacterized protein from the budding yeast Saccharomyces cerevisiae with a full length of 67 amino acids. The complete amino acid sequence is MNKRYKLYRVWYYYAHQTVCITSTGFALCFVVQAKTAGLGVTPITSLYGDKKEHLGKLLVPLVLYQI . As an uncharacterized protein, its specific biological function remains to be elucidated, creating opportunities for novel research directions. The protein can be recombinantly expressed with an N-terminal His-tag in E. coli expression systems, facilitating purification and subsequent functional studies .
Current structural analyses suggest the protein may contain functional domains that could provide insights into its role in yeast cellular processes. Researchers should approach this protein as a potential component in unidentified pathways, particularly considering the location of its encoding gene within the S. cerevisiae genome and potential regulatory elements surrounding it.
To predict potential functions of this uncharacterized protein, researchers should implement a multi-layered bioinformatic approach:
Sequence-based analysis: Begin with BLAST and PSI-BLAST searches against protein databases to identify distant homologs. Follow with multiple sequence alignment to identify conserved residues that might indicate functional importance.
Structural prediction: Employ tools like AlphaFold, I-TASSER, or Phyre2 to predict tertiary structure, which may reveal structural similarities to proteins of known function despite low sequence identity.
Domain prediction: Use InterProScan, SMART, and Pfam to identify potential functional domains, motifs, or signatures.
Evolutionary analysis: Perform phylogenetic analysis to understand evolutionary relationships and potential functional conservation across species.
Co-expression network analysis: Analyze gene expression datasets to identify genes co-expressed with YDR182W-A, which may suggest functional relationships.
Protein-protein interaction prediction: Use tools like STRING database to predict potential interacting partners based on various evidence types .
The lack of characterized pathways involving YDR182W-A in the current literature necessitates these comprehensive approaches to generate testable hypotheses about its function .
Based on available data, E. coli has been successfully used as an expression system for recombinant YDR182W-A production . The methodology typically involves:
Construct design: The gene encoding YDR182W-A should be codon-optimized for E. coli expression and cloned into an appropriate expression vector containing an N-terminal His-tag for purification purposes.
Expression conditions: Optimal conditions often include:
E. coli strain selection (BL21(DE3), Rosetta, or Arctic Express for potentially challenging proteins)
Induction with IPTG at concentrations between 0.1-1.0 mM
Expression at lower temperatures (16-25°C) to enhance proper folding
Extended expression time (16-24 hours) for maximum yield
Alternative systems: If E. coli yields improperly folded protein, consider:
Yeast expression systems (particularly S. cerevisiae or Pichia pastoris) which may provide more native folding conditions
Cell-free expression systems for rapid screening of conditions
Mammalian cell expression for complex folding requirements
The Creative BioMart products utilize E. coli as the expression host with high purity (>90% as determined by SDS-PAGE) , suggesting this is a viable approach for research quantities.
Efficient purification of His-tagged YDR182W-A requires a methodical approach:
Cell lysis optimization:
Buffer composition (typically phosphate or Tris-based buffers, pH 7.5-8.0)
Inclusion of appropriate protease inhibitors
Mild detergents if the protein has hydrophobic regions
Sonication or high-pressure homogenization parameters
Immobilized metal affinity chromatography (IMAC):
Ni-NTA or Co-based resins for His-tag binding
Optimization of imidazole concentration in wash buffers (10-40 mM) to reduce non-specific binding
Gradient or step elution protocols with increasing imidazole (100-500 mM)
Secondary purification:
Size exclusion chromatography to remove aggregates and ensure monodispersity
Ion exchange chromatography if charge-based separation would improve purity
Quality control:
SDS-PAGE analysis with Coomassie or silver staining
Western blot with anti-His antibodies
Mass spectrometry for identity confirmation
Storage recommendations indicate avoiding repeated freeze-thaw cycles, with working aliquots stored at 4°C for up to one week . Long-term storage should be at -20°C in a stabilizing buffer containing glycerol or other cryoprotectants.
To identify protein-protein interactions involving YDR182W-A, researchers should employ a combination of complementary techniques:
Affinity purification coupled with mass spectrometry (AP-MS):
Express tagged YDR182W-A in S. cerevisiae
Pull-down experiments using the His-tag
Identify co-precipitated proteins by mass spectrometry
Validate with reciprocal tagging of candidate interactors
Yeast two-hybrid (Y2H) screening:
Proximity-based labeling:
BioID or APEX2 fusion proteins to identify proximal proteins in vivo
Particularly useful for transient or weak interactions
Synthetic genetic array (SGA) analysis:
Cross-linking mass spectrometry (XL-MS):
Chemical cross-linking to capture direct protein-protein contacts
Structural insights into interaction interfaces
Surface plasmon resonance (SPR) or bio-layer interferometry (BLI):
For quantitative binding kinetics of candidate interactions
The TORC2-Ypk1 signaling pathway analysis described in search result 3 provides a methodological template for how such interaction studies might be structured, even though it does not directly mention YDR182W-A .
Determining the subcellular localization of YDR182W-A is crucial for understanding its function. Researchers should consider these methodological approaches:
Fluorescent protein tagging:
C-terminal or N-terminal GFP/mCherry fusion constructs
Expression from native promoter to maintain physiological levels
Live-cell imaging under various growth conditions
Co-localization with known organelle markers
Immunofluorescence microscopy:
Generation of specific antibodies against YDR182W-A
Fixation and permeabilization optimization for yeast cells
Double labeling with organelle markers
Biochemical fractionation:
Differential centrifugation to separate cellular compartments
Western blot analysis of fractions using anti-His antibodies
Comparison with known compartment markers
Proximity-dependent biotinylation:
BioID or APEX2 fusion for in vivo proximity labeling
Identification of biotinylated proteins to infer localization
Computational prediction:
Analysis of protein sequence for localization signals
Tools like PSORT, TargetP, and DeepLoc for prediction
Electron microscopy:
Immunogold labeling for high-resolution localization
Correlative light and electron microscopy for functional context
The knowledge that YDR182W-A can be expressed as a recombinant protein with a His-tag suggests that epitope tagging approaches are feasible and likely to yield meaningful results.
While YDR182W-A's function remains uncharacterized, researchers can investigate its potential involvement in yeast metabolic pathways through systematic approaches:
Transcriptional co-regulation analysis:
Examine expression profiles under various conditions
Identify co-regulated genes with known pathway associations
Analyze promoter regions for common regulatory elements
Metabolomic profiling:
Compare metabolite profiles between wild-type and YDR182W-A deletion/overexpression strains
Focus on changes in key metabolic intermediates
Use stable isotope labeling to track metabolic flux
Integration with existing pathway models:
Growth phenotype analysis:
Systematic testing of growth under various carbon sources, stress conditions
Chemical genomics approaches with inhibitors of known pathways
Flux balance analysis:
The systematic assessment of variables that was used in engineering S. cerevisiae for β-ionone production represents a methodological template that could be adapted to study YDR182W-A's potential metabolic functions.
Comprehensive genetic analysis can provide significant insights into YDR182W-A function:
Gene deletion and overexpression studies:
Creation of YDR182W-A knockout strains
Controlled overexpression using inducible promoters
Phenotypic characterization under various conditions
Synthetic genetic array analysis:
CRISPR-Cas9 genome editing:
Suppressor screens:
Identify mutations that suppress phenotypes of YDR182W-A deletion/overexpression
Second-site suppressors can reveal pathway connections
Transcriptional response analysis:
RNA-seq to identify genes affected by YDR182W-A manipulation
ChIP-seq if YDR182W-A has potential DNA-binding domains
Protein domain analysis:
Construction of chimeric proteins
Deletion of specific protein regions to identify functional domains
The chemical genetic approach described in search result 3, which scores deleterious effects from overexpression when a kinase of interest is inhibited , could be particularly valuable for investigating YDR182W-A's potential involvement in signaling pathways.
Determining the structure of a small, uncharacterized protein like YDR182W-A (67 amino acids) requires careful selection of appropriate techniques:
Given YDR182W-A's small size, NMR spectroscopy would likely be the most appropriate experimental technique, providing both structural information and insights into potential conformational dynamics.
Functional annotation of uncharacterized proteins presents significant challenges that require integrated approaches:
Multi-omics integration:
Combine transcriptomics, proteomics, metabolomics, and phenomics data
Network-based approaches to predict function through guilt by association
Machine learning algorithms to identify patterns across multiple data types
Activity-based protein profiling:
Chemical probes to detect specific enzymatic activities
Metabolite affinity purification to identify binding partners
Systematic screening against metabolite libraries
Comparative genomics across yeast species:
Analysis of YDR182W-A orthologs in other fungi
Investigation of gene neighborhood conservation
Identification of correlated gene loss/gain patterns
High-throughput phenotyping:
Automated microscopy to detect subtle morphological changes
Growth measurements under thousands of conditions
Chemogenomic profiling with diverse chemical libraries
CRISPR-based functional genomics:
Genome-wide screens to identify genetic interactions
CRISPRi/CRISPRa for modulating expression levels
Domain-focused mutagenesis to map functional regions
Systems biology modeling:
The systematic, multi-tiered approach described in search result 3 for identifying kinase substrates demonstrates how combining chemical genetics, bioinformatics, and biochemical validation can successfully annotate previously unknown functions .
Investigating the regulation of YDR182W-A expression requires careful experimental design:
Promoter analysis:
Transcriptional analysis:
qRT-PCR for targeted quantification under various conditions
RNA-seq for genome-wide expression context
Single-cell RNA-seq to detect population heterogeneity
CAGE-seq or similar techniques to precisely map transcription start sites
Translation regulation:
Ribosome profiling to assess translational efficiency
Analysis of 5' and 3' UTR sequences for regulatory elements
Investigation of potential upstream open reading frames (uORFs)
Epigenetic regulation:
ChIP-seq for histone modifications near the YDR182W-A locus
DNA methylation analysis if relevant to the study system
Chromatin accessibility assays (ATAC-seq, DNase-seq)
Environmental response profiling:
Systematic analysis of YDR182W-A expression under:
Different carbon sources
Stress conditions (oxidative, osmotic, temperature)
Growth phases
Nutrient limitations
Integration with signaling pathways:
The approach used in the β-carotene and β-ionone production study, which systematically optimized gene expression through careful manipulation of transcriptional unit architecture, integration sites, and gene dosage , provides a methodological framework that could be adapted for studying YDR182W-A regulation.