Protein:
Immunohistochemistry (IHC): Used to detect C3orf36 in formalin-fixed tissues at dilutions of 1:100–500 .
Antibody Competition: Recombinant antigens (e.g., Novus Biologicals) validate antibody specificity in blocking assays .
Cancer Research: Chromosome 3p25.3 deletions are common in cancers . C3orf36’s association with apoptosis mediators (RASSF1) and angiogenesis suppressors (SEMA3B) positions it as a potential biomarker .
Therapeutic Insights:
Limitations: Functional studies using recombinant C3orf36 (e.g., Antibodies-online’s ABIN3090675) remain exploratory .
C3orf36 (Chromosome 3 Open Reading Frame 36) is characterized as a putative immune regulator believed to play a significant role in modulating immune responses and maintaining immune homeostasis. Current research suggests potential connections between this protein and various pathological conditions including cancer development, autoimmune disorders, and inflammatory conditions . The protein is encoded by the C3orf36 gene (Gene ID: 80111) and has UniProt accession code Q3SXR2 . As research into this protein continues to evolve, understanding its functional characteristics provides critical context for antibody-based detection methods in experimental systems.
Currently available C3orf36 antibodies are predominantly rabbit-derived polyclonal antibodies that target specific regions of the human C3orf36 protein. Most commercial antibodies target the C-terminal region, specifically amino acids within positions 116-165 of the human C3orf36 protein (NP_079317) . These antibodies are typically immunoaffinity purified and available in unconjugated formats, though some suppliers offer conjugated versions with biotin, FITC, or HRP for specialized applications . The polyclonal nature of these antibodies provides researchers with broad epitope recognition capabilities, which can be advantageous for detecting proteins that may undergo post-translational modifications or exist in different conformational states.
Current C3orf36 antibodies have been validated primarily for Western blot (WB) applications , with some additionally validated for immunohistochemistry (IHC) and ELISA techniques . For Western blot applications, these antibodies effectively detect denatured C3orf36 protein from human samples. Immunohistochemistry applications have been specifically demonstrated with paraffin-embedded human tissues, including melanoma samples at dilutions of approximately 1:100 . For optimal experimental outcomes, researchers should follow supplier-recommended dilution ranges, which typically include 1:2000-1:10000 for ELISA and 1:20-1:200 for IHC applications .
Validation of C3orf36 antibody specificity requires multiple complementary approaches. First, researchers should include appropriate positive controls (tissues or cell lines known to express C3orf36) and negative controls (tissues lacking C3orf36 expression) in each experiment. Second, employing blocking peptides specifically designed for C3orf36 antibodies can confirm binding specificity by competitively inhibiting antibody-antigen interactions . Third, results from antibody-based detection should be corroborated with orthogonal methods such as mRNA expression analysis. Finally, comparing results from multiple antibodies targeting different epitopes of C3orf36 can provide additional confidence in specificity. BLAST analysis indicates that the current antibodies recognize sequences with 100% identity to human C3orf36, suggesting high species specificity .
Detecting low-abundance C3orf36 protein requires systematic optimization of experimental parameters. Begin by implementing antigen retrieval methods appropriate for the sample type; for formalin-fixed tissues, citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) heating protocols may improve epitope accessibility. Signal amplification systems such as avidin-biotin complex (ABC) or tyramide signal amplification (TSA) can significantly enhance detection sensitivity without increasing background. When performing Western blot analysis of low-abundance samples, extended transfer times (overnight at lower voltage) combined with high-sensitivity chemiluminescent substrates can improve detection limits. Additionally, sample enrichment through immunoprecipitation prior to Western blotting can concentrate C3orf36 protein. For each methodological adjustment, parallel positive controls at different dilutions should be processed to establish a calibration curve for quantification.
Cross-reactivity challenges with C3orf36 antibodies can be addressed through multiple technical approaches. First, implement a tiered antibody dilution series to identify the optimal concentration that maximizes specific signal while minimizing non-specific binding. Pre-adsorption of the antibody with the immunizing peptide can effectively reduce cross-reactivity by occupying non-specific binding sites. For Western blot applications, increasing the stringency of wash steps (higher salt concentration or mild detergents) and optimizing blocking conditions (5% BSA may be superior to milk-based blockers for certain applications) can significantly reduce background. If cross-reactivity persists, consider computational modeling approaches to predict potential cross-reactive epitopes based on sequence homology, as outlined in recent antibody specificity research . This biophysics-informed modeling can help identify binding modes associated with specific or cross-reactive interactions.
The impact of fixation chemistry on C3orf36 epitope preservation creates significant methodological considerations. Formaldehyde fixation forms methylene bridges that can mask C-terminal epitopes critical for antibody recognition, potentially requiring extended antigen retrieval protocols. In contrast, acetone or methanol fixation preserves protein conformation differently by precipitating proteins without extensive cross-linking. Researchers should systematically compare multiple fixation protocols when establishing C3orf36 detection methods for new tissue types. For optimal results, limit fixation duration to the minimum required for structural preservation (typically 24-48 hours for formaldehyde), as extended fixation exponentially increases epitope masking. When working with cell cultures, mild permeabilization with 0.1-0.3% Triton X-100 often provides optimal epitope accessibility while maintaining cellular architecture. Each parameter modification should be documented with standardized positive controls to establish reproducible protocols.
Successful multiplexing of C3orf36 detection with other biomarkers requires careful consideration of antibody compatibility and detection system parameters. First, select antibodies from different host species (e.g., rabbit anti-C3orf36 with mouse anti-secondary marker) to enable species-specific secondary antibody discrimination. For multi-color fluorescence applications, ensure spectral separation between fluorophores to minimize bleed-through (typically >50nm between emission maxima). When using enzyme-based detection systems, sequential rather than simultaneous development often provides superior discrimination. Consider tyramide signal amplification for sequential multiplexing, as it allows antibody stripping while preserving the amplified signal from previous detection rounds. For each new antibody combination, perform single-staining controls alongside multiplexed samples to verify that antibody performance is not compromised in the multiplexed environment. This systematic approach ensures reliable co-localization data for C3orf36 with other markers of interest.
Investigating C3orf36 protein interactions requires a multi-method experimental design strategy. Begin with co-immunoprecipitation using anti-C3orf36 antibodies followed by mass spectrometry to identify potential binding partners without prior assumptions. This approach should be complemented with proximity ligation assays (PLA) for in situ verification of identified interactions within cellular contexts. For detailed binding kinetics, surface plasmon resonance (SPR) or biolayer interferometry using purified C3orf36 protein can provide association and dissociation constants. When expressing recombinant C3orf36 for interaction studies, consider both N-terminal and C-terminal tagging strategies, as the positioning of tags may differentially impact protein interactions. Computational prediction of potential binding interfaces based on protein structure can guide mutation studies to validate specific interaction domains. For each identified interaction, reciprocal co-immunoprecipitation (pulling down with antibodies against both C3orf36 and the partner protein) provides stronger evidence of physiologically relevant associations.
Functional neutralization studies with C3orf36 antibodies require rigorous controls to establish specificity and physiological relevance. Essential controls include: (1) Isotype-matched control antibodies from the same species to distinguish specific from non-specific effects; (2) Dose-response titrations to establish the minimal effective concentration for neutralization; (3) Pre-adsorption controls using immunizing peptides to confirm specificity; (4) Parallel knockdown or knockout models (siRNA or CRISPR-based) to corroborate antibody-based neutralization effects; (5) Rescue experiments where recombinant C3orf36 is added back to neutralized systems to reverse observed effects. When interpreting results, researchers should assess the kinetics of neutralization effects relative to the known half-life of C3orf36 protein. For in vivo neutralization studies, biodistribution analysis of the antibody should be performed to confirm access to relevant tissue compartments where C3orf36 functions.
Advanced computational methods can significantly enhance C3orf36 antibody design through biophysics-informed modeling approaches. Begin by implementing epitope prediction algorithms that integrate protein structure, surface accessibility, and hydrophilicity profiles to identify optimal target regions. Machine learning models trained on phage display selection data can predict binding affinities across sequence variants, enabling in silico screening before experimental validation . These models can disentangle multiple binding modes associated with specific ligands, allowing researchers to design antibodies with customized specificity profiles – either highly specific for C3orf36 or with controlled cross-reactivity to related proteins . By incorporating structural information into the computational pipeline, researchers can predict conformational epitopes beyond linear sequences. Molecular dynamics simulations can further refine predictions by modeling the flexibility of target epitopes and their accessibility under physiological conditions. This integrated computational-experimental approach can accelerate the development of next-generation C3orf36 antibodies with precisely engineered specificity profiles.
Accurate quantification of C3orf36 across diverse tissues requires standardized methodological approaches to overcome tissue-specific variables. First, establish a panel of reference tissues with validated C3orf36 expression levels to serve as inter-experimental calibrators. For immunohistochemical quantification, digital image analysis using standardized acquisition parameters and automated thresholding algorithms provides superior reproducibility compared to manual scoring. When comparing across tissue types, normalize C3orf36 signals to tissue-appropriate housekeeping proteins that maintain stable expression in the specific contexts under investigation. Consider tissue-specific protein extraction efficiencies; fibrous tissues may require modified extraction buffers containing higher detergent concentrations or mechanical disruption methods. For absolute quantification, generate a standard curve using recombinant C3orf36 protein spiked into a matrix that mimics the target tissue's composition. Finally, triangulate protein-level measurements with transcript analysis through RT-qPCR or RNA-seq to distinguish between transcriptional and post-transcriptional regulation of C3orf36 expression.
Inconsistent results across platforms often stem from platform-specific variables that affect antibody performance. First, conduct systematic epitope mapping to identify whether the recognized epitopes are equally accessible in different experimental contexts (native vs. denatured conditions). For each platform, optimize sample preparation independently, as fixation requirements for IHC differ fundamentally from preparation for Western blotting. When transitioning between applications, adjust antibody concentrations based on the effective epitope concentration in each system rather than using a standard dilution across all methods. Consider buffer composition effects; the presence of detergents, reducing agents, or metal ions may differentially impact epitope-antibody interactions across platforms. Document lot-to-lot variations by maintaining reference samples tested with each new antibody lot. Finally, when possible, deploy multiple antibodies targeting different epitopes of C3orf36 to distinguish true biological variation from technical artifacts.
Validating C3orf36 knockout or knockdown models requires a multi-level verification strategy. At the genomic level, PCR-based genotyping with primers flanking the targeted region can confirm genetic modifications, while Sanger sequencing verifies the precise nature of the mutation. For transcript analysis, design qPCR primers targeting multiple exons, including those outside the targeted region, to detect potential splice variants or truncated transcripts that might retain partial functionality. At the protein level, use multiple antibodies recognizing different epitopes of C3orf36 to ensure comprehensive detection of any residual protein expression. Western blot analysis should include extended exposure times to detect low-abundance truncated proteins. Functional validation is equally critical; measure known downstream effects of C3orf36 activity to confirm that the knockout/knockdown produces expected functional consequences. Finally, rescue experiments where wild-type C3orf36 is reintroduced should reverse the observed phenotypes, providing definitive evidence that the effects are specifically attributable to C3orf36 depletion rather than off-target effects.
Discrepancies between protein and mRNA levels of C3orf36 require systematic investigation of multiple biological and technical factors. First, examine the temporal relationship between measurements, as protein expression typically lags behind mRNA changes, creating apparent discordance in dynamic systems. Second, assess post-transcriptional regulation mechanisms including microRNA-mediated repression, which may suppress translation without affecting mRNA levels. Third, investigate protein stability through cycloheximide chase experiments to determine if extended protein half-life maintains C3orf36 protein levels despite reduced transcript levels. Fourth, evaluate tissue-specific translational efficiency, which can vary substantially between cell types. From a technical perspective, verify primer specificity for RT-qPCR and probe for alternatively spliced variants that might escape detection with exon-specific primers. Similarly, confirm antibody specificity through knockout validation controls. When discrepancies persist, consider implementing ribosome profiling to directly measure translational activity of C3orf36 mRNA, providing an intermediate measurement between transcript abundance and protein levels.
Investigating C3orf36's role in disease pathology requires strategic application of antibodies across multiple experimental paradigms. For cancer research, tissue microarray analysis with anti-C3orf36 antibodies can establish expression patterns across tumor stages and correlate with patient outcomes. In inflammatory conditions, dual immunofluorescence for C3orf36 and inflammatory cell markers can reveal potential regulatory interactions at sites of inflammation. For mechanistic studies, chromatin immunoprecipitation sequencing (ChIP-seq) using antibodies against transcription factors predicted to regulate C3orf36 can elucidate expression control in disease states. Patient-derived samples should be analyzed with standardized protocols that incorporate matched control tissues processed identically. When extending to animal models of disease, ensure that the antibody cross-reactivity with the model organism's C3orf36 homolog has been experimentally verified. Finally, therapeutic potential can be explored through antibody-mediated neutralization in relevant disease models, comparing outcomes with genetic ablation approaches to distinguish direct from indirect effects.
Developing C3orf36 antibodies with precisely tailored specificity requires integration of biophysical modeling with experimental validation. Begin by conducting comprehensive epitope mapping of the C3orf36 protein to identify regions that are either unique or shared with related proteins. Implement phage display selections against multiple ligands simultaneously to identify antibody variants with diverse binding profiles . Apply computational modeling that incorporates binding modes for each potential target to predict sequences with desired specificity characteristics . This approach can generate antibodies with either high specificity for a particular epitope or controlled cross-reactivity to multiple targets . For therapeutic applications where discrimination between closely related epitopes is critical, integrate negative selection steps against structurally similar but functionally distinct targets. Validate engineered specificity through comprehensive cross-reactivity testing against predicted off-target proteins based on sequence homology. This integrated approach leverages both computational prediction and experimental selection to achieve precisely controlled antibody specificity not possible through traditional immunization methods alone.
Integration of C3orf36 antibody detection with single-cell technologies enables unprecedented resolution of expression heterogeneity within complex tissues. For mass cytometry (CyTOF) applications, metal-conjugated anti-C3orf36 antibodies can be incorporated into multi-parameter panels alongside lineage and functional markers to correlate C3orf36 expression with cellular phenotypes. When designing single-cell protocols, optimize fixation and permeabilization conditions specifically for C3orf36 detection, as overly harsh permeabilization may extract cytoplasmic proteins while insufficient permeabilization limits antibody access. For imaging mass cytometry or multiplexed ion beam imaging, validate the performance of C3orf36 antibodies in the presence of the complete antibody panel to identify potential steric hindrances. Single-cell RNA-seq data can be integrated with protein-level measurements through CITE-seq approaches, where oligonucleotide-tagged antibodies enable simultaneous protein and transcript profiling. This multi-modal profiling can reveal post-transcriptional regulation mechanisms specific to C3orf36. For spatial transcriptomics integration, optimize tissue preparation protocols that preserve both RNA integrity and protein epitopes for dual detection of C3orf36 transcript and protein within spatial contexts.