C5orf22 Antibody is a research tool targeting the C5orf22 protein, a poorly characterized gene product encoded by the C5orf22 gene on human chromosome 5 (5p13.3). While the protein’s precise function remains elusive, it is hypothesized to participate in DNA splicing complexes, with emerging roles in cancer pathogenesis and muscular dystrophy . The antibody enables detection and functional studies of C5orf22 via techniques such as Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) .
Cancer Research: Upregulated C5orf22 correlates with reduced survival in breast cancer patients .
Muscular Dystrophy: Reduced expression observed in tibial muscular dystrophy .
Cardiomyopathy: Increased expression linked to non-ischemic cardiomyopathy .
C5orf22 is implicated in the WBP11/PQBP1 splicing complex, regulating genes involved in DNA repair and immunomodulation . Knockdown experiments revealed:
Aberrant Splicing: Disrupted alternative splicing events in genes critical for cell cycle regulation .
Protein Interactions: Over 20 interactors identified, including WBP11, OSM, and Surf2 .
C5orf22 (Chromosome 5 Open Reading Frame 22) is a human protein encoded by a gene located on chromosome 5. While its precise function remains under investigation, it represents one of many uncharacterized proteins (UPFs) that require further study to elucidate their biological roles. Researchers investigate C5orf22 using specific antibodies to understand its expression patterns, subcellular localization, and potential functions in normal and disease states .
Multiple C5orf22 antibody types exist for research applications:
| Antibody Type | Host | Target Epitopes | Conjugations | Applications |
|---|---|---|---|---|
| Polyclonal | Rabbit | AA 179-228, AA 101-420, AA 301-400, AA 153-202 | Unconjugated, FITC, HRP, Biotin, AbBy Fluor® 555/680 | WB, ELISA, IHC, IF, FACS, ICC |
| Polyclonal | Rabbit | AA 179-228 | Alexa Fluor® 700 | IHC-P |
Most commonly used are rabbit polyclonal antibodies targeting various epitopes of the human C5orf22 protein, with unconjugated versions being predominant for western blotting applications .
C5orf22 antibodies are typically produced using one of two approaches:
Synthetic peptide immunization: Antibodies like ABIN6750628 are generated using synthetic peptides corresponding to specific regions of human C5orf22 (e.g., aa179-228). These peptides are conjugated to carrier proteins, injected into host animals (commonly rabbits), and the resulting antibodies are purified through immunoaffinity methods .
Recombinant protein immunization: Some antibodies are produced using larger recombinant fragments of the C5orf22 protein as immunogens. For example, antibody orb354187 was generated using recombinant human C5orf22 protein (101-420AA) .
Both methods produce polyclonal antibodies with different binding characteristics and applications.
For rigorous validation of C5orf22 antibodies, implement this multi-step approach:
Genetic validation (gold standard): Test antibodies by immunoblot comparing parental cell lines to CRISPR/Cas9-generated C5orf22 knockout (KO) lines. Valid antibodies should show robust signal in parental cells that is absent or significantly reduced in KO cells .
Orthogonal validation: Compare antibody-based detection with mRNA expression data or results from multiple antibodies targeting different epitopes of C5orf22 .
Immunoprecipitation followed by mass spectrometry: This confirms the antibody's ability to enrich for C5orf22 and identifies potential cross-reacting proteins .
Overexpression systems: Test antibody response to exogenously expressed tagged C5orf22 (e.g., Myc-DYKDDDDK tagged C5orf22) .
This comprehensive validation strategy ensures antibody specificity for the intended target and appropriate application .
To reduce non-specific binding of C5orf22 antibodies:
Optimize blocking conditions: Test different blocking agents (5% BSA, 5% non-fat milk, commercial blockers) to identify optimal blocking for your specific sample type and antibody.
Titrate antibody concentration: Perform dilution series experiments to identify the minimum concentration that provides specific signal. For C5orf22 antibodies, recommended dilutions typically range from 1:200-1:500 for IHC and 1:250-1:2000 for WB, but optimization is necessary for each experimental system .
Modify washing protocols: Implement more stringent washing procedures, including increased wash duration, volume, or detergent concentration.
Include appropriate controls: Always include negative controls (C5orf22 knockout or knockdown samples) to distinguish between specific and non-specific signals .
Pre-adsorption: Consider pre-adsorbing the antibody with the immunizing peptide to confirm specificity of observed signals.
Knockout validation represents the most rigorous method for antibody specificity determination for several reasons:
Unbiased validation: CRISPR/Cas9-mediated knockout provides an isogenic control lacking only the target antigen, allowing direct assessment of antibody specificity against the endogenous protein .
Application versatility: Unlike other validation methods, genetic approaches are suitable for validating antibodies across all applications (WB, IHC, IF, etc.) .
Historical limitations overcome: Previous antibody validation was hampered by lack of suitable controls, but modern gene editing techniques now make knockout-based validation feasible and accessible .
Reveals problematic antibodies: Studies with C9ORF72 antibodies demonstrated that some widely used antibodies failed knockout validation despite being used in highly cited papers, raising concerns about previously reported findings. This same principle applies to C5orf22 antibody research .
For C5orf22 antibodies, implementation of knockout validation helps prevent publication of potentially misleading results based on non-specific antibody reactions.
For optimal Western blot results with C5orf22 antibodies:
Sample preparation:
Use RIPA or 1% Triton X-100 containing buffers for efficient protein extraction
Load 50μg of total protein per lane
Use 5-16% gradient gels for optimal resolution of C5orf22 (~49 kDa)
Transfer conditions:
Transfer to nitrocellulose membranes (preferred over PVDF for C5orf22)
Verify transfer efficiency with Ponceau S staining
Blocking and antibody incubation:
Detection:
For quantitative analysis, use fluorescently-labeled secondary antibodies and imaging systems like LI-COR Odyssey
For enhanced sensitivity, use HRP-conjugated secondaries with enhanced chemiluminescence
Controls:
This methodology has been optimized based on published protocols for C5orf22 and related protein research.
For selecting the optimal C5orf22 antibody for IHC applications:
Validation status: Prioritize antibodies like HPA043062 that have been validated by the Human Protein Atlas project through extensive tissue microarray testing on 44 normal human tissues and 20 common cancer types .
Epitope considerations: Select antibodies targeting epitopes that remain accessible after fixation and antigen retrieval. For C5orf22, antibodies targeting the middle region (e.g., AA 179-228) often perform well in IHC applications .
Fixation compatibility: Confirm the antibody is compatible with your fixation method (formalin-fixed paraffin-embedded vs. frozen sections). Some C5orf22 antibodies like those marked for "IHC (fro)" and "IHC (p)" are validated for both frozen and paraffin sections .
Visualization format: For brightfield microscopy, use unconjugated primary antibodies with appropriate secondary detection systems. For fluorescence applications, consider directly conjugated antibodies like NBP2-98180AF700 (Alexa Fluor® 700 conjugated) .
Dilution optimization: Always optimize antibody dilution for your specific tissue and conditions. For C5orf22 antibodies, recommended starting dilutions are typically 1:200-1:500 for IHC .
Based on protein expression databases and experimental evidence, these cell lines serve as appropriate positive controls for C5orf22 antibody testing:
HEK-293 cells: Human embryonic kidney cells show detectable expression of C5orf22 and have been used successfully for recombinant C5orf22 expression, making them suitable for both endogenous and overexpression studies .
MCF7 cells: Human breast cancer cell line with documented C5orf22 expression, used for Western blot validation of some commercial C5orf22 antibodies .
PaxDB-recommended lines: Reference the PaxDB proteomics database to identify cell lines with high C5orf22 expression, following the approach used for antibody validation pipelines .
When selecting positive control cell lines, consider:
Expression level of the target protein
Compatibility with your experimental system
Availability of isogenic knockout cells for validation
Cell type relevance to your research question
As shown in antibody validation studies, using cell lines with relatively high target protein expression improves sensitivity and specificity assessment .
When facing weak or absent signal with C5orf22 antibodies, systematically address these potential issues:
Protein expression levels:
Confirm C5orf22 expression in your sample using proteomics databases or RT-PCR
Consider using cell lines with known higher expression of C5orf22
For very low abundance, implement enrichment strategies (e.g., immunoprecipitation before detection)
Antibody selection issues:
Test alternative C5orf22 antibodies targeting different epitopes
Verify antibody functionality using positive controls
Check antibody storage and handling conditions (avoid repeated freeze-thaw cycles)
Protocol optimization:
Increase antibody concentration (within manufacturer's recommended range)
Extend primary antibody incubation time (overnight at 4°C)
Optimize antigen retrieval methods for IHC/IF
For Western blots, try different protein extraction buffers to improve solubilization
Detection system limitations:
Use more sensitive detection systems (e.g., enhanced chemiluminescence for WB)
Consider signal amplification techniques (e.g., tyramide signal amplification for IHC)
For fluorescent applications, select fluorophores with minimal background in your sample type
Sample preparation concerns:
Ensure protein denaturation is complete for WB applications
Add protease inhibitors to prevent protein degradation
Check protein extraction efficiency with positive control proteins
Implementing this systematic approach will help identify and address specific causes of weak signal when working with C5orf22 antibodies.
To reduce high background when using C5orf22 antibodies in immunofluorescence applications:
Fixation and permeabilization optimization:
Test different fixation methods (4% PFA, methanol, or acetone)
Adjust permeabilization conditions (concentration and duration of detergent exposure)
Include a quenching step to reduce autofluorescence (0.1-1% sodium borohydride or 50mM NH₄Cl)
Blocking improvements:
Extend blocking time (1-2 hours at room temperature)
Test different blocking agents (normal serum from secondary antibody host species, BSA, casein, commercial blockers)
Add 0.1-0.3% Triton X-100 to blocking buffer to reduce non-specific membrane binding
Antibody dilution and incubation:
Washing protocol enhancements:
Increase number and duration of wash steps
Use PBS-T (PBS + 0.1% Tween-20) for more effective washing
Perform final washes with plain PBS to remove detergent
Controls and counterstaining:
Include secondary-only controls to assess non-specific secondary binding
Use appropriate counterstains that don't interfere with your signal
Consider using directly conjugated antibodies to eliminate secondary antibody background
These optimizations should significantly improve signal-to-noise ratio when working with C5orf22 antibodies in immunofluorescence applications.
To distinguish between technical variation and biological heterogeneity when working with C5orf22 antibodies:
Implement comprehensive controls:
Positive controls (samples with known C5orf22 expression)
Negative controls (C5orf22 knockout samples)
Internal controls (detection of housekeeping proteins in the same samples)
Technical replicates (repeated analysis of the same sample)
Biological replicates (multiple independent samples from the same condition)
Quantitative assessment:
Use quantitative detection methods (e.g., LI-COR Odyssey system for WB)
Calculate coefficients of variation for technical replicates (<15% typically indicates acceptable technical reproducibility)
Apply appropriate statistical tests to determine significance of observed differences
Multi-antibody approach:
Test multiple antibodies targeting different C5orf22 epitopes
Compare results from antibodies from different manufacturers/sources
Assess correlation between different antibody results (high correlation suggests biological signal rather than technical artifacts)
Orthogonal validation:
Compare antibody-based results with mRNA expression data
Validate key findings with non-antibody methods (e.g., mass spectrometry)
Consider using tagged C5orf22 constructs for expression validation
Documentation and standardization:
Maintain detailed records of all experimental conditions
Standardize protocols to minimize technical variation
Document antibody lot numbers, as lot-to-lot variation can significantly impact results
This systematic approach helps differentiate between technical noise and genuine biological variation in C5orf22 detection.
For successful co-immunoprecipitation (co-IP) experiments to identify C5orf22 interacting partners:
Antibody selection for co-IP:
Based on immunoprecipitation validation strategies used for similar proteins like C9ORF72, select antibodies that effectively precipitate endogenous C5orf22
Pre-test candidate antibodies by performing small-scale IPs followed by Western blot to assess enrichment efficiency
Identify antibodies that can capture at least 50-70% of C5orf22 from lysates for optimal results
Buffer optimization:
Use mild lysis conditions to preserve protein-protein interactions (e.g., 1% NP-40 or 0.5% CHAPS buffer)
Include protease and phosphatase inhibitors
Consider crosslinking agents for transient interactions
Test multiple buffer conditions to optimize interaction preservation while maintaining antibody binding
Experimental procedure:
Pre-couple antibodies to protein A/G beads (1-5 μg antibody per IP)
Prepare detergent-solubilized lysates (1-2 mg total protein per IP)
Include appropriate controls:
Beads alone (no antibody)
Isotype control antibody
Lysate from C5orf22 knockout cells
Interaction analysis:
Perform mass spectrometry analysis of immunoprecipitates to identify potential binding partners
Confirm interactions by reciprocal co-IP or other interaction methods
Validate specificity by comparing proteins identified in C5orf22 IP versus control IPs
Focus on proteins enriched in C5orf22 IP from wild-type cells but absent in IPs from knockout cells
Result interpretation:
Prioritize proteins consistently identified across technical replicates
Consider functional categories and pathway analysis of identified partners
Compare partners to known interactors of proteins with similar subcellular localization or function
This approach mirrors successful strategies used to identify interaction partners for other uncharacterized proteins .
For accurate quantitative analysis of C5orf22 expression across tissues or cell types:
Antibody validation requirements:
Select antibodies validated in knockout/knockdown systems to ensure specificity
Verify linear detection range using titrated recombinant C5orf22 proteins
Confirm equal detection efficiency across different sample types
Assess potential interference from tissue-specific post-translational modifications
Technical approach selection:
For tissue analysis: Choose between IHC with digital image analysis or Western blotting of tissue lysates
For cell-type specific analysis: Consider flow cytometry (if surface exposed) or single-cell imaging approaches
For absolute quantification: Implement approaches using recombinant C5orf22 protein standards
Quantitative detection optimization:
For Western blotting: Use fluorescently-labeled secondary antibodies and imaging systems like LI-COR Odyssey
For IHC: Apply consistent staining protocols and calibrated image acquisition settings
Include standard curves when possible
Process all samples in parallel to minimize batch effects
Normalization strategy:
Select appropriate housekeeping proteins as loading controls
Consider using total protein normalization (e.g., Ponceau S staining)
For tissue analysis, normalize to tissue-specific reference proteins rather than ubiquitous housekeeping proteins
Validate normalization approach for each tissue/cell type
Statistical analysis:
Apply appropriate statistical tests based on data distribution
Account for biological variability between replicates
Consider multiple testing correction for comparisons across many tissues/cell types
Present data with appropriate measures of central tendency and dispersion
Following these guidelines ensures reliable quantitative comparisons of C5orf22 expression across diverse biological samples.
Active learning strategies can significantly enhance C5orf22 antibody research efficiency:
Epitope selection optimization:
Apply active learning algorithms to predict optimal antigenic regions of C5orf22 for antibody development
Iteratively test and refine epitope selection based on antibody performance feedback
This approach can reduce the number of required antigen variants by up to 35% compared to traditional methods
Iterative antibody validation pipeline:
Start with a small set of validation experiments on candidate antibodies
Use results to inform which additional experiments would be most informative
Prioritize subsequent testing based on preliminary specificity assessments
This reduces resource expenditure on extensively validating poor candidates
Application-specific optimization:
Begin with base protocol conditions
Design intelligent sampling of parameter space rather than exhaustive grid searches
Use Bayesian optimization to suggest next experimental conditions based on previous results
This approaches optimal conditions more rapidly than traditional one-factor-at-a-time optimization
Cross-reactivity assessment:
Apply active learning to efficiently identify potential cross-reactive proteins
Test against predicted cross-reactive targets rather than random proteins
Reduce testing burden while maintaining confidence in specificity assessments
Library-on-library screening approaches:
For developing new C5orf22 antibodies, implement active learning strategies to guide antibody-antigen pairing experiments
This can speed up the learning process by approximately 28 steps compared to random sampling approaches
Focus resources on the most informative experiments to rapidly converge on optimal antibodies
By integrating these active learning approaches, researchers can achieve more efficient and effective results in C5orf22 antibody development and application while minimizing experimental resources.
A multi-technique approach using C5orf22 antibodies provides complementary insights:
Integrated workflow example:
Begin with Western blotting to confirm expression and molecular weight in tissues/cells of interest
Follow with immunofluorescence to determine subcellular localization
Perform IHC to examine tissue distribution patterns
Use immunoprecipitation-mass spectrometry to identify interaction partners
Apply proximity ligation assays to confirm specific interactions in situ
Technique complementarity benefits:
Biochemical techniques (WB, IP) provide quantitative data about expression levels and interactions
Imaging techniques (IF, IHC) provide spatial information about distribution and co-localization
Functional assays (e.g., antibody-mediated inhibition) connect molecular presence to biological activity
Validation through cross-technique confirmation:
Verify C5orf22 subcellular localization through both biochemical fractionation+WB and immunofluorescence
Confirm protein-protein interactions through both co-IP and proximity ligation assays
Compare tissue expression patterns between IHC and tissue Western blots
Data integration strategy:
Develop unified models incorporating expression, localization, and interaction data
Use bioinformatic approaches to integrate antibody-derived data with transcriptomic and proteomic datasets
Identify discrepancies between techniques as opportunities for biological discovery
This comprehensive approach has demonstrated success in characterizing previously uncharacterized proteins like C9ORF72, revealing its localization to phagosomes/lysosomes .
To address limitations in conventional C5orf22 antibody applications:
Proximity-dependent labeling approaches:
Express C5orf22 fused to BioID or TurboID to identify proximal proteins
This overcomes limitations in co-IP by capturing transient interactions and spatial proximity
Allows identification of the C5orf22 microenvironment without relying on stable interactions
Single-molecule localization microscopy:
Apply super-resolution techniques (STORM, PALM) using validated C5orf22 antibodies
Achieve 10-20nm resolution compared to ~250nm in conventional microscopy
Provides precise nanoscale localization of C5orf22 relative to cellular structures
Single-cell antibody-based proteomics:
Implement mass cytometry (CyTOF) using metal-conjugated C5orf22 antibodies
Measure C5orf22 expression alongside dozens of other proteins at single-cell resolution
Reveals cell-to-cell heterogeneity in expression and correlation with cellular states
Spatially-resolved proteomics:
Apply imaging mass cytometry or multiplexed ion beam imaging (MIBI)
Maintain tissue architecture while measuring C5orf22 distribution
Correlate with numerous other proteins in the same tissue section
Live-cell nanobody applications:
Develop nanobodies against C5orf22 that function in intracellular environments
Express as fusion proteins with fluorescent reporters for live-cell imaging
Track dynamics and trafficking of C5orf22 in real-time without fixation artifacts
These advanced approaches extend beyond limitations of traditional antibody applications while building upon the specificity verification principles established through rigorous antibody validation protocols .
CRISPR-based technologies are transforming protein detection and localization studies, offering alternatives and complements to traditional C5orf22 antibody approaches:
Endogenous protein tagging:
Use CRISPR-Cas9 to knock-in fluorescent tags or epitope tags at the C5orf22 locus
Provides visualization or pulldown of C5orf22 at endogenous expression levels
Overcomes antibody specificity concerns while maintaining native regulation
Example advantage: Tagged C5orf22 can be tracked in live cells without fixation artifacts
CUT&TAG and CUT&RUN technologies:
For studying C5orf22 DNA/chromatin interactions (if relevant)
Uses CRISPR-guided targeting rather than traditional ChIP-seq
Offers higher sensitivity and lower background than antibody-based ChIP approaches
Potentially applicable if C5orf22 has nuclear functions
CRISPR interference/activation systems:
Deploy CRISPRi/a to modulate C5orf22 expression
Creates calibrated knockdown or overexpression systems for antibody validation
Generates graded expression levels to test antibody sensitivity and linearity
More precise than traditional siRNA approaches
Programmable RNA-guided protein manipulation:
Adapt CRISPR systems to target C5orf22 protein directly
Enables acute protein degradation, relocalization, or modification
Provides temporal control not possible with genetic knockout approaches
Complements antibody detection by creating controlled experimental perturbations
Integration with antibody-based approaches:
Use CRISPR knockout cells as definitive negative controls for antibody validation
Combine CRISPR-based perturbation with antibody-based detection for functional studies
Deploy CRISPR screens to identify genes affecting C5orf22 expression, localization, or function
Validate antibody-identified interactions through CRISPR perturbation of interaction partners