The FITC-conjugated antibody is optimized for detecting C22orf39 in:
Cultured Cells (IF-cc): Localizes C22orf39 in intracellular compartments.
Paraffin-Embedded Sections (IF-p): Identifies protein expression in fixed tissues.
Immunocytochemistry (ICC): Used in cell-based assays to study subcellular distribution .
Method | Dilution Range |
---|---|
IF (cc/p) | 1:50–200 |
ICC | 1:50–200 |
Target Specificity: Binds to the AA 1-142 region of human C22orf39 .
Cross-Reactivity: Limited to human samples (no cross-reactivity reported in other species) .
Gene Name: Chromosome 22 Open Reading Frame 39.
Function: The exact role of C22orf39 remains under investigation, but it is associated with chromosomal regulation and may play a role in cellular processes requiring precise protein localization .
Fluorescence Detection: Enables real-time visualization under fluorescence microscopy.
Sensitivity: FITC’s green emission (excitation: 495 nm, emission: 519 nm) enhances signal-to-noise ratio in IF/ICC .
Format | Conjugate | Key Applications | Catalog Examples |
---|---|---|---|
Unconjugated | – | ELISA, IHC, WB | ABIN7175041 |
FITC | FITC | IF, ICC | ABIN7175043, CSB-PA761294LC01HU |
HRP | HRP | WB, ELISA | ABIN2171313 |
Biotin | Biotin | ELISA, IHC | ABIN7175044 |
AbBy Fluor® 594 | AbBy Fluor® 594 | IF (p), WB | ABIN2802309 |
C22ORF39 (Chromosome 22 Open Reading Frame 39) is a human protein encoded by a gene located on chromosome 22. Research into this protein is facilitated through the use of specific antibodies that can detect its presence in various cellular contexts. While the function of C22ORF39 remains under investigation, antibodies against this target enable researchers to study its expression patterns, localization, and potential involvement in cellular processes. Antibodies with fluorescent conjugates like FITC allow for direct visualization of this protein in microscopy and flow cytometry applications .
C22ORF39 antibodies are available with specificity to different amino acid regions of the protein, most commonly AA 1-142 and AA 31-105. These different targeting specificities reflect antibodies raised against distinct epitopes within the C22ORF39 protein. The antibody targeting AA 31-105 recognizes a more restricted region of the protein, which may provide higher specificity for certain applications. In contrast, the antibody targeting AA 1-142 covers a broader region, potentially offering greater sensitivity but possibly increased risk of cross-reactivity. The choice between these depends on the specific research question and application requirements .
FITC (Fluorescein Isothiocyanate) conjugation provides a bright green fluorescence when excited with appropriate wavelengths (typically ~495 nm), making it useful for direct visualization without secondary antibodies. Compared to other conjugates like HRP (used primarily for colorimetric detection in Western blotting and ELISA) or biotin (requiring an additional streptavidin step), FITC-conjugated antibodies allow for direct one-step detection in fluorescence-based applications. This makes FITC conjugates particularly valuable for immunofluorescence microscopy and flow cytometry applications where direct visualization is required .
Based on the available information, C22ORF39 antibodies are typically produced in rabbit hosts and are polyclonal in nature. Polyclonal antibodies offer the advantage of recognizing multiple epitopes within the target protein, potentially providing stronger signal amplification compared to monoclonal antibodies. The rabbit origin makes these antibodies compatible with many experimental systems, particularly when working with human samples, as they minimize cross-reactivity issues that might occur with mouse-derived antibodies when studying human tissues .
When designing experiments with C22ORF39 FITC-conjugated antibodies, several controls are essential: (1) Negative controls using isotype-matched FITC-conjugated IgG from the same host species (rabbit) to establish background fluorescence levels; (2) Positive controls using cells or tissues known to express C22ORF39; (3) Blocking peptide controls where the antibody is pre-incubated with the immunizing peptide to confirm specificity; and (4) Secondary antibody-only controls when using indirect detection methods. These controls help validate results and distinguish specific from non-specific signals, which is particularly important in fluorescence-based applications where autofluorescence can be problematic .
Optimization for C22ORF39 antibody staining in immunofluorescence requires careful attention to several parameters. Begin with fixation method testing (4% paraformaldehyde versus methanol) as this can significantly impact epitope accessibility. Optimize antibody concentration through a titration series (typically 1:50 to 1:500) to determine the dilution providing maximum specific signal with minimal background. Incubation conditions should be evaluated, with overnight incubation at 4°C often yielding better results than shorter incubations at room temperature. Additionally, test different blocking solutions (5-10% normal serum or BSA) to minimize non-specific binding. Finally, consider permeabilization methods when targeting intracellular epitopes, testing different concentrations of Triton X-100 or saponin .
Based on immunofluorescence protocols, C22ORF39 antibodies are generally compatible with standard fixation methods. Cells and tissue sections can be fixed prior to antibody staining and then incubated with the primary antibody overnight at 4°C. The most common fixation approach involves using 4% paraformaldehyde, which preserves cellular architecture while maintaining antigen accessibility. Alternative fixation methods like methanol or acetone might be suitable for certain applications but should be empirically tested as they can affect FITC fluorescence intensity or epitope availability. The fixation duration should be optimized to balance structural preservation with antibody accessibility to the target .
Quantification of C22ORF39 expression using FITC-conjugated antibodies can be achieved through multiple approaches. For flow cytometry, direct measurement of FITC fluorescence intensity correlates with protein expression levels, allowing for population-based quantification. In microscopy applications, fluorescence intensity can be measured using software like ImageJ, calculating integrated density values from defined regions of interest. For more precise quantification, techniques like quantitative immunofluorescence using standard curves of recombinant proteins can be employed. When comparing expression across samples, it's essential to normalize fluorescence intensity to an appropriate housekeeping protein or use ratiometric approaches to account for variations in cell number and imaging conditions .
The optimal immunofluorescence protocol for C22ORF39 FITC-conjugated antibodies follows these methodological steps: (1) Fix specimens using 4% paraformaldehyde for 15-20 minutes at room temperature; (2) Wash 3 times with PBS; (3) If detecting intracellular targets, permeabilize with 0.1-0.5% Triton X-100 for 10 minutes; (4) Block with 5-10% normal serum in PBS for 1 hour at room temperature; (5) Incubate with diluted FITC-conjugated C22ORF39 antibody (typically 1:100 to 1:500) overnight at 4°C in a humidified chamber; (6) Wash 3 times with PBS; (7) Counterstain nuclei with DAPI; (8) Mount with anti-fade mounting medium. For optimal results, protect the slides from light during and after the antibody incubation to prevent photobleaching of the FITC fluorophore .
Yes, C22ORF39 FITC-conjugated antibodies can be effectively used in flow cytometry applications. For optimal results, cells should be fixed with 2-4% paraformaldehyde and permeabilized if detecting intracellular epitopes. A typical protocol involves: (1) Harvesting and washing cells in flow cytometry buffer; (2) Fixing with 2-4% paraformaldehyde for 10-15 minutes; (3) Permeabilizing with 0.1% saponin or Triton X-100 if needed; (4) Blocking with 2-5% BSA or FBS; (5) Incubating with diluted FITC-conjugated C22ORF39 antibody (1:50 to 1:200) for 30-60 minutes at 4°C; (6) Washing to remove unbound antibody; (7) Analyzing on a flow cytometer with appropriate excitation (488 nm laser) and emission filters for FITC detection. This approach allows for quantitative assessment of C22ORF39 expression at the single-cell level .
FITC-conjugated C22ORF39 antibodies offer several methodological advantages over unconjugated alternatives: (1) Single-step detection eliminates the need for secondary antibodies, reducing protocol time and potential background; (2) Reduced cross-reactivity issues since no secondary antibody is required; (3) Direct quantification of signal intensity correlating with protein expression; (4) Compatibility with multi-color immunofluorescence when combined with antibodies conjugated to spectrally distinct fluorophores; (5) Reliable signal localization without displacement concerns that can occur with secondary antibody binding. These advantages make FITC-conjugated antibodies particularly valuable for applications requiring high-resolution imaging or precise quantification of protein expression levels .
When designing multi-color immunofluorescence experiments with C22ORF39 FITC-conjugated antibodies, careful selection of complementary fluorophores is essential to minimize spectral overlap. Ideal combinations include: (1) FITC (green) with TRITC/Cy3 (red) and DAPI (blue); (2) FITC with far-red fluorophores like Cy5 or Alexa Fluor 647; or (3) FITC with Pacific Blue and Alexa Fluor 555. When combining multiple fluorophores, sequential staining may be necessary to prevent antibody cross-reactivity. Additionally, proper instrument setup with appropriate excitation sources and emission filters is critical, along with careful compensation settings when using flow cytometry. For microscopy applications, sequential image acquisition can minimize bleed-through between channels .
The relationship between signaling pathway activation and C22ORF39 detection requires sophisticated experimental design. Although direct evidence for C22ORF39 phosphorylation is limited, potential interactions with AKT and ERK1/2 signaling pathways may influence its detection. Researchers investigating these relationships should employ dual staining approaches using C22ORF39 FITC-conjugated antibodies alongside phospho-specific antibodies against AKT (pAKT) or ERK1/2 (pERK1/2). Treatment with pathway inhibitors such as U0126 (ERK1/2 inhibitor) or LY294002 (PI3K/AKT inhibitor) prior to staining can reveal dynamic relationships between pathway activation and C22ORF39 expression or localization. Quantitative co-localization analysis and stimulation experiments with growth factors may further elucidate these regulatory relationships .
Several advanced microscopy techniques can significantly enhance spatial resolution when using C22ORF39 FITC-conjugated antibodies: (1) Confocal microscopy eliminates out-of-focus light, providing optical sectioning with ~200 nm lateral resolution; (2) Super-resolution techniques such as Structured Illumination Microscopy (SIM) can achieve ~100 nm resolution with standard FITC labels; (3) Stimulated Emission Depletion (STED) microscopy can push resolution to ~50 nm but may require more photostable fluorophores than FITC; (4) Single Molecule Localization Microscopy (STORM/PALM) offers ~20 nm resolution but typically requires specialized fluorophores with specific photoswitching properties. When implementing these techniques, sample preparation becomes increasingly critical, with attention to fixation quality, antibody concentration optimization, and mounting media selection to minimize spherical aberrations and maximize signal-to-noise ratios .
Quantitative co-localization analysis between C22ORF39 and other cellular components requires rigorous methodological approaches: (1) Dual immunofluorescence staining using C22ORF39 FITC-conjugated antibody and a spectrally distinct fluorophore-conjugated antibody against the protein of interest; (2) High-quality confocal imaging with appropriate controls for bleed-through; (3) Analysis using specialized software (ImageJ with Coloc2 plugin, CellProfiler, or similar) to calculate Pearson's correlation coefficient, Mander's overlap coefficient, or intensity correlation quotient; (4) Threshold determination through objective methods such as Costes' approach; (5) Statistical validation through comparison with randomized images. For dynamic co-localization studies, live cell imaging using photoactivatable or photoconvertible fluorescent proteins fused to proteins of interest can be combined with immunofluorescence for the endogenous C22ORF39 protein after fixation .
Optimizing fixation protocols requires balancing epitope preservation with fluorophore stability. For FITC-conjugated C22ORF39 antibodies, consider these methodological adjustments: (1) Test mild fixation with 2-4% paraformaldehyde for 10-15 minutes at room temperature, which typically preserves most epitopes while maintaining FITC fluorescence; (2) Avoid prolonged fixation which can mask epitopes through excessive cross-linking; (3) If signal is weak, try antigen retrieval methods such as citrate buffer (pH 6.0) heating or enzymatic retrieval with proteinase K; (4) For difficult-to-detect epitopes, consider dual fixation approaches using brief glutaraldehyde (0.05-0.1%) followed by paraformaldehyde; (5) Post-fixation quenching with glycine (100mM) or ammonium chloride can reduce autofluorescence from aldehyde groups. Always prepare controls with varying fixation conditions to determine the optimal protocol for specific applications .
High background with FITC-conjugated C22ORF39 antibodies can result from several factors: (1) Insufficient blocking—extend blocking time to 1-2 hours with 5-10% serum or BSA; (2) Excessive antibody concentration—perform titration experiments to determine optimal dilution; (3) Non-specific binding—add 0.1-0.3% Triton X-100 or 0.05% Tween-20 to washing buffers; (4) Autofluorescence—treat samples with sodium borohydride (1mg/ml) for 10 minutes or use Sudan Black B (0.1-0.3%) to quench lipofuscin fluorescence; (5) Inadequate washing—increase wash duration and number of washes; (6) Suboptimal fixation causing protein precipitation—optimize fixation protocols; (7) Cross-reactivity with endogenous Fc receptors—add Fc receptor blocking reagents to blocking buffer. Systematic evaluation of these factors can significantly improve signal-to-noise ratios in immunofluorescence applications .
When encountering weak or absent signals with C22ORF39 FITC-conjugated antibodies, implement a systematic troubleshooting approach: (1) Verify antibody functionality with positive control samples known to express the target; (2) Optimize antibody concentration through titration experiments; (3) Modify fixation conditions as epitope accessibility may be compromised; (4) Implement antigen retrieval techniques such as heat-induced epitope retrieval with citrate buffer (pH 6.0); (5) Extend primary antibody incubation to overnight at 4°C to increase binding opportunity; (6) Evaluate permeabilization efficiency, potentially increasing detergent concentration for intracellular targets; (7) Consider signal amplification systems such as tyramide signal amplification; (8) Check for fluorophore degradation by testing a new antibody batch or alternative conjugate; (9) Optimize microscope settings including exposure time, gain, and laser power to detect weak signals .
Proper normalization is critical for accurate quantification of C22ORF39 expression across experimental conditions. Recommended methodological approaches include: (1) Internal control normalization using housekeeping proteins such as β-actin or GAPDH, ideally with a spectrally distinct fluorophore conjugate; (2) Cell number normalization through nuclear counterstaining with DAPI and calculating per-cell expression values; (3) Total protein normalization using general protein stains like Coomassie blue or Ponceau S for parallel Western blot validation; (4) Reference sample normalization where all experimental conditions are compared to a standard reference sample run in parallel; (5) For flow cytometry, using median fluorescence intensity rather than mean values to minimize the effect of outliers. Statistical analysis should include tests for normal distribution of data before applying parametric statistical methods .
Distinguishing between protein levels and post-translational modifications requires specialized approaches: (1) Dual staining with antibodies specific to unmodified versus modified forms of C22ORF39, using spectrally distinct fluorophores; (2) Comparison of staining patterns between total protein (using antibodies recognizing all forms) and modified protein (using modification-specific antibodies); (3) Treatment with enzymes that remove specific modifications (phosphatases for phosphorylation, deubiquitinases for ubiquitination) prior to staining; (4) Correlation of immunofluorescence data with Western blot analysis using modification-specific antibodies; (5) Pharmacological manipulation of relevant pathways controlling modifications, such as kinase inhibitors for phosphorylation. These approaches can reveal subtle regulatory mechanisms affecting C22ORF39 function beyond simple expression level changes .
Flow cytometry data for C22ORF39 expression requires appropriate statistical analysis: (1) For comparing expression between two populations, use non-parametric tests like Mann-Whitney U test, as fluorescence intensity distributions are often non-normal; (2) For multiple group comparisons, apply Kruskal-Wallis with post-hoc tests like Dunn's multiple comparison; (3) Use robust measures like median fluorescence intensity rather than mean values; (4) For bimodal distributions indicating subpopulations, apply mixture modeling or clustering algorithms; (5) For correlating C22ORF39 expression with other cellular parameters, use Spearman's rank correlation; (6) When analyzing time-course experiments, consider repeated measures ANOVA or mixed effects models; (7) Always report effect sizes alongside p-values to indicate biological significance. Sample size calculations should be performed during experimental design to ensure adequate statistical power for detecting anticipated differences .
Integrating C22ORF39 FITC-conjugated antibodies into multiparameter flow cytometry requires careful panel design: (1) Position FITC in a channel with minimal spillover into other detectors, typically FL1 on standard cytometers; (2) Select compatible fluorophores with minimal spectral overlap, such as PE, APC, and Pacific Blue; (3) Perform comprehensive compensation using single-stained controls for each fluorophore; (4) Include Fluorescence Minus One (FMO) controls to establish accurate gating boundaries; (5) When studying rare cell populations, consider using brighter fluorophores than FITC for those markers; (6) Optimize antibody concentrations individually before combining in the final panel; (7) For panels exceeding 5-6 colors, consider spectral cytometry platforms with advanced unmixing algorithms. This methodological approach ensures accurate identification of cell populations expressing C22ORF39 in relation to other markers of interest .
Studying subcellular localization of C22ORF39 requires high-resolution imaging approaches: (1) Confocal microscopy with z-stack acquisition to capture the three-dimensional distribution; (2) Co-staining with established organelle markers (e.g., DAPI for nucleus, MitoTracker for mitochondria, ER-Tracker for endoplasmic reticulum) using spectrally distinct fluorophores; (3) Super-resolution microscopy techniques like Airyscan or Structured Illumination for enhanced spatial resolution; (4) Quantitative co-localization analysis using specialized software to calculate Pearson's or Mander's coefficients; (5) Cellular fractionation followed by Western blotting to validate microscopy findings; (6) Live-cell imaging using cell-permeable nuclear or organelle stains combined with immunofluorescence after fixation. These approaches provide complementary information about the spatial distribution of C22ORF39 within cells under various conditions .
Studying protein-protein interactions involving C22ORF39 can be approached through several methodologies: (1) Proximity Ligation Assay (PLA) combining C22ORF39 FITC-conjugated antibody with an antibody against the potential interacting partner; (2) Co-immunoprecipitation followed by Western blot validation; (3) Fluorescence Resonance Energy Transfer (FRET) between FITC (donor) and a suitable acceptor fluorophore attached to the interaction partner; (4) High-resolution confocal microscopy with quantitative co-localization analysis; (5) Live-cell imaging combined with techniques like Fluorescence Recovery After Photobleaching (FRAP) or Fluorescence Loss In Photobleaching (FLIP) to assess dynamic interactions; (6) In situ Proximity Ligation Assay for detecting interactions with sensitivity to detect single-molecule events. These approaches provide complementary information about potential interaction partners and the dynamics of these interactions in cellular contexts .