KEGG: gga:770642
UniGene: Gga.9355
The UPF0542 protein C5orf43 homolog (RCJMB04_3o3) from Gallus gallus (Chicken) is a small protein with 74 amino acids in its expression region. Its complete amino acid sequence is: MFDVKAWAVYIVEWAAKDPYGFLTTVILVLTPLFIISAALSWKLAKMIETREREQKKKRKRQENIVKAKRAKKD. The protein has been assigned the UniProt accession number Q5F409 and is classified as part of the UPF0542 protein family .
To characterize this protein thoroughly, researchers should employ multiple complementary approaches:
Primary structure analysis using mass spectrometry
Secondary structure prediction using circular dichroism spectroscopy
Tertiary structure determination using X-ray crystallography or NMR spectroscopy
Functional domain identification through bioinformatic analysis of conserved regions
Evaluate protein stability at various temperatures (-80°C, -20°C, 4°C) using activity assays at defined time intervals
Test buffer composition effects by examining:
pH ranges (5.0-9.0)
Salt concentrations (0-500 mM NaCl)
Additives (glycerol percentages from 10-50%)
Conduct freeze-thaw cycle testing (1, 3, 5, and 10 cycles)
Perform aggregation analysis using dynamic light scattering after various storage periods
The results should be presented as a stability matrix showing relative activity retention across all tested conditions.
Selection of an optimal expression system for Recombinant UPF0542 protein C5orf43 homolog depends on research objectives and required protein characteristics. While bacterial expression in E. coli is commonly employed for recombinant proteins , researchers should consider several factors:
Expression System | Advantages | Limitations | Recommended Applications |
---|---|---|---|
E. coli | High yield, rapid growth, cost-effective | Limited post-translational modifications, potential inclusion bodies | Structural studies, antibody generation |
Insect cells | Eukaryotic PTMs, proper folding | More complex, moderate yield | Functional studies requiring native conformation |
Mammalian cells | Full range of PTMs, native folding | Costly, lower yield | Interaction studies, functional assays |
Cell-free systems | Rapid, avoids toxicity issues | Expensive, limited scale | Protein engineering, directed evolution |
For initial characterization studies, E. coli expression with optimization of induction parameters (IPTG concentration, temperature, and induction time) is often most practical. For the UPF0542 protein specifically, expression optimization should focus on solubility enhancement through fusion tags and modified induction protocols (e.g., using 0.5-1.0 mM IPTG at 25°C for 16 hours) .
Triple resonance NMR techniques offer powerful approaches for studying protein structure and interactions at atomic resolution. While traditional NMR approaches can be challenging for certain proteins, recent advancements in triple resonance experiments provide enhanced sensitivity and resolution.
To apply triple resonance NMR to UPF0542 protein studies:
Sample preparation:
Express 13C/15N double-labeled protein in minimal media
Purify to >95% homogeneity
Prepare 0.5-1.0 mM protein in NMR buffer (typically 50 mM phosphate, 50-150 mM NaCl, pH 6.5-7.5)
Experiment selection:
Analysis workflow:
Backbone assignment using established protocols
Secondary structure determination from chemical shift indices
NOE-based distance restraints for tertiary structure
Relaxation measurements for dynamics analysis
Triple resonance approaches can be particularly valuable for detecting subtle conformational changes upon ligand binding or mutation, providing insights into UPF0542 protein function that would be inaccessible through other structural techniques .
Designing chimeric constructs with UPF0542 protein domains requires careful consideration of multiple factors to ensure proper expression and functionality:
Domain boundary identification:
Analyze sequence conservation across species
Use structure prediction algorithms to identify independently folding units
Consider hydrophobic core preservation when selecting fusion points
Linker design strategies:
Flexible linkers (GGGGS)n for independent domain function
Rigid linkers (EAAAK)n when domain orientation is critical
Specific cleavage sites for post-expression processing
Expression optimization considerations:
Construct validation approaches:
For UPF0542 protein specifically, the intact protein is relatively small (74 amino acids), making it potentially suitable as a fusion partner. When designing chimeric constructs, the secondary structure prediction using SOPMA and 3D structure generation using Swiss Model server should be performed to ensure proper domain folding .
Resolving contradictory results in protein localization studies requires systematic investigation using complementary approaches:
Methodological reconciliation strategy:
Compare fixation protocols (paraformaldehyde vs. methanol)
Assess antibody specificity through Western blot analysis
Evaluate expression level effects (physiological vs. overexpression)
Consider cell type-specific differences in localization machinery
Multi-technique verification approach:
Technique | Resolution | Live/Fixed | Advantages | Limitations |
---|---|---|---|---|
Immunofluorescence | ~200 nm | Fixed | Multiple protein detection | Fixation artifacts |
Fluorescent protein fusion | ~200 nm | Live | Dynamic studies | Tag interference |
Biochemical fractionation | N/A | Fixed | Quantitative | Poor spatial resolution |
Proximity labeling | ~10 nm | Live | In situ neighbors | Non-specific labeling |
Super-resolution microscopy | 10-50 nm | Both | High resolution | Complex analysis |
Computational analysis of protein sequence:
Predict localization signals (NLS, NES, signal peptides)
Analyze post-translational modification sites affecting localization
Examine interaction partners with known localizations
Design of definitive experiments:
CRISPR/Cas9 endogenous tagging to maintain physiological expression
Split fluorescent protein complementation to confirm interaction-dependent localization
Inducible expression systems to monitor localization kinetics
When addressing UPF0542 protein localization specifically, sequence analysis suggests potential membrane association based on the presence of hydrophobic regions within its amino acid sequence (MFDVKAWAVYIVEWAAKDPYGFLTTVILVLTPLFIISAALSWKLAKMIETREREQKKKRKRQENIVKAKRAKKD) , which should be experimentally verified.
Developing an optimal purification strategy for Recombinant UPF0542 protein requires a multi-step approach that balances purity, yield, and functional integrity:
Initial capture phase:
Immobilized Metal Affinity Chromatography (IMAC) using Ni-NTA agarose is recommended for His-tagged UPF0542 protein
Optimize binding conditions: test various imidazole concentrations (5-20 mM) in binding buffer to reduce non-specific binding
For denaturing conditions, use 8M urea or 6M guanidine-HCl buffer systems
For native conditions, include mild detergents (0.1% Triton X-100) if membrane association is suspected
Intermediate purification:
Ion exchange chromatography (IEX) based on theoretical pI calculation from amino acid sequence
Size exclusion chromatography (SEC) to remove aggregates and separate oligomeric states
Polishing and validation:
Scale-up considerations:
Implement tangential flow filtration for concentration rather than centrifugal concentrators
Monitor and minimize endotoxin levels for downstream biological applications
Validate batch consistency through activity assays and analytical SEC
The purification protocol should be optimized iteratively, with full documentation of recovery and purity at each step to identify bottlenecks in the process.
Designing controlled experiments to elucidate UPF0542 protein function requires a comprehensive approach:
Comparative genomics foundation:
Analyze conservation across species
Identify co-evolved gene clusters
Examine expression patterns in different tissues/conditions
Loss-of-function experimental design:
CRISPR/Cas9 knockout with phenotypic characterization
RNAi-mediated knockdown with dose-response assessment
Dominant-negative mutant expression
Gain-of-function experimental design:
Controlled overexpression using inducible promoters
Rescue experiments in knockout backgrounds
Ectopic expression in heterologous systems
Interaction mapping strategy:
Yeast two-hybrid screening with appropriate controls
Co-immunoprecipitation followed by mass spectrometry
Proximity labeling (BioID or APEX) in relevant cellular compartments
Essential controls for rigorous interpretation:
Empty vector controls for all expression experiments
Scrambled siRNA/sgRNA controls for knockdown/knockout
Inactive mutant controls (e.g., predicted active site mutations)
Cell viability assessments to rule out non-specific toxicity
Time-course experiments to distinguish primary from secondary effects
For the UPF0542 protein specifically, its small size (74 amino acids) and sequence characteristics suggest potential roles in protein-protein interactions or membrane-associated functions that should be systematically tested through these controlled experimental designs.
Analysis of post-translational modifications (PTMs) on UPF0542 protein requires an integrated workflow combining enrichment, detection, and quantification strategies:
Comprehensive PTM mapping strategy:
PTM Type | Enrichment Method | Detection Technique | Quantification Approach |
---|---|---|---|
Phosphorylation | TiO2/IMAC | LC-MS/MS, Phos-tag gels | SILAC, TMT labeling |
Glycosylation | Lectin affinity | PNGase F/mass shift | Glycopeptide MS/MS |
Ubiquitination | K-ε-GG antibodies | Ubiquitin remnant MS | Spectral counting |
Acetylation | Anti-acetyl lysine | MS/MS, Western blot | Label-free quantification |
Methylation | Anti-methyl antibodies | High-resolution MS | Heavy methyl SILAC |
Site-specific analysis workflow:
In silico prediction of modification sites using algorithms specific to each PTM
Site-directed mutagenesis of predicted sites to confirm functional relevance
Generation of site-specific antibodies for high-throughput monitoring
Parallel reaction monitoring (PRM) for targeted MS analysis of modified peptides
Temporal dynamics assessment:
Pulse-chase experiments with metabolic labeling
Time-resolved proteomics following stimulation
In vitro enzyme assays with purified modification enzymes
Functional impact determination:
Correlation of modification status with protein activity
Structural analysis of modified vs. unmodified protein
Interactome analysis conditioned on modification state
Given the small size of UPF0542 protein and its sequence characteristics, particular attention should be paid to potential phosphorylation sites within the C-terminal region containing multiple lysine residues (KLAKMIETREREQKKKRKRQENIVKAKRAKKD) , which may regulate its interactions or localization.
Optimizing CRISPR/Cas9 technology for UPF0542 protein function studies in avian systems requires addressing several unique challenges:
Delivery optimization strategy:
Viral vectors: Lentivirus for cell lines, adeno-associated virus for in vivo applications
Non-viral approaches: Lipofection for cell lines, electroporation for primary cells
In ovo injection techniques for developmental studies
Guide RNA design considerations:
Target sequence specificity verification against chicken genome
Evaluation of guide efficiency using prediction algorithms
Design of homology-directed repair templates for knock-in experiments
Off-target analysis specific to Gallus gallus genome
Validation protocol development:
T7 Endonuclease I assay for initial editing efficiency assessment
Deep sequencing to quantify editing precision and off-target effects
RT-qPCR and Western blot to confirm knockdown at RNA and protein levels
Phenotypic characterization using standardized assays
Experimental design for functional studies:
Generation of cell line panels (knockout, knockdown, domain deletions)
Complementation studies with wild-type and mutant constructs
Integration with transcriptomic and proteomic profiling
Developmental timing considerations for in vivo studies
Given the limited information available specifically for UPF0542 protein C5orf43 homolog, comparative analysis with mammalian orthologs can guide hypothesis generation for functional studies in avian systems.
Predicting UPF0542 protein interaction partners requires integration of multiple computational approaches:
Sequence-based prediction methods:
Motif identification for known interaction domains
Conservation analysis to identify co-evolved residues
Machine learning approaches trained on known protein-protein interactions
Structure-based prediction techniques:
Homology modeling followed by molecular docking
Binding site analysis using fpocket or SiteMap
Molecular dynamics simulations to assess interaction stability
Interface residue prediction using PIER or ProMate
Network-based approaches:
Gene co-expression analysis across tissues and conditions
Phylogenetic profiling to identify functionally related proteins
Literature-based relationship extraction using NLP
Integration of high-throughput interaction data from related species
Validation strategy development:
Design of targeted experiments to confirm high-confidence predictions
Cross-validation using orthogonal computational methods
Sensitivity analysis to assess prediction robustness
For UPF0542 protein specifically, analysis of its amino acid sequence (MFDVKAWAVYIVEWAAKDPYGFLTTVILVLTPLFIISAALSWKLAKMIETREREQKKKRKRQENIVKAKRAKKD) suggests potential for membrane association and protein-protein interactions through its lysine-rich C-terminal region, which should guide computational prediction parameters.
Addressing solubility challenges in Recombinant UPF0542 protein expression requires a systematic troubleshooting approach:
Expression condition optimization:
Construct modification strategies:
Fusion tags selection matrix:
Fusion Tag | Size | Effect on Solubility | Purification Method |
---|---|---|---|
MBP | 42 kDa | High enhancement | Amylose resin |
SUMO | 11 kDa | Moderate enhancement | IMAC |
Thioredoxin | 12 kDa | Moderate enhancement | IMAC |
GST | 26 kDa | Variable | Glutathione resin |
NusA | 55 kDa | High enhancement | IMAC |
Codon optimization for Gallus gallus-derived protein
Truncation constructs to identify soluble domains
Site-directed mutagenesis of hydrophobic residues
Extraction condition optimization:
Detergent screening (non-ionic: Triton X-100, NP-40; zwitterionic: CHAPS)
Ionic strength variation (100-500 mM NaCl)
pH optimization (6.0-9.0) based on theoretical pI
Addition of stabilizing co-factors or ligands
Refolding strategy development (if inclusion bodies are unavoidable):
On-column refolding during IMAC purification
Dilution refolding with redox pair (GSH/GSSG)
Dialysis-based refolding with decreasing denaturant gradient
Chaperone-assisted refolding
Given the small size of UPF0542 protein (74 amino acids) and its sequence characteristics, particular attention should be paid to the hydrophobic regions that may contribute to aggregation during expression .
Overcoming detection limitations in protein interaction studies requires implementation of sensitive techniques and proper experimental design:
Enhanced detection methodology selection:
Proximity ligation assay (PLA) for in situ interaction detection
FRET/BRET for real-time interaction monitoring
Single-molecule pull-down for detection of low-abundance complexes
Crosslinking mass spectrometry for transient interaction capture
Signal amplification approaches:
Tyramide signal amplification for immunodetection
Rolling circle amplification for proximity assays
Multi-epitope tagging for enhanced antibody recognition
Click chemistry-based labeling for improved signal-to-noise ratio
Enrichment strategy optimization:
Tandem affinity purification for complex stability
Size exclusion chromatography for native complex isolation
Density gradient fractionation for compartment-specific interactions
Immunoprecipitation under optimized buffer conditions
Control implementation for result validation:
Interaction-deficient mutants as negative controls
Known interaction partners as positive controls
Competition assays with excess unlabeled protein
Reciprocal tagging to confirm bidirectional pull-down
For UPF0542 protein specifically, its small size may present detection challenges that can be addressed through specialized approaches such as chemical crosslinking prior to purification or the use of split reporter systems (luciferase complementation assay, split-GFP) to enhance signal detection.