The PRAMEF10 antibody, biotin conjugated, targets the PRAME family member 10 protein (UniProt: O60809), which functions as a transcriptional repressor inhibiting retinoic acid receptor (RAR) signaling. This inhibition blocks retinoic acid-mediated cellular processes such as differentiation and apoptosis, making PRAMEF10 relevant in cancer and developmental biology research .
Biotin conjugation enhances detection sensitivity by enabling streptavidin-mediated signal amplification in assays like ELISA, Western blotting, and immunofluorescence .
Note: Biotin amplification (e.g., tyramide-based systems) further enhances sensitivity in low-expression scenarios .
Retinoic Acid Signaling: PRAMEF10 suppresses RAR-mediated transcription, promoting cell proliferation in melanoma and other cancers .
Protein Interaction Mapping: Proximity labeling techniques (e.g., BioID) using biotinylated antibodies identified PRAMEF10-associated NE proteins like SLAP75 and SAMP1, revealing its role in nuclear envelope organization .
Clinical Relevance: Overexpression correlates with poor prognosis in malignancies, suggesting utility as a diagnostic biomarker .
Biotin Interference: Endogenous biotin in tissues (e.g., liver, kidney) causes false positives, necessitating blocking protocols .
Stability: ProClin 300 preservative is toxic, requiring careful handling .
Cross-Reactivity: No data available for non-human models, limiting translational studies .
What is PRAMEF10 and why is it studied in research?
PRAMEF10 (PRAME Family Member 10) belongs to the PRAME gene family, which has garnered scientific interest primarily in cancer biology and immunology research. The protein is typically studied to understand its biological functions in normal cellular processes and its potential role in disease states. While the specific functions of PRAMEF10 remain under investigation, researchers often examine its expression patterns across different tissues and its potential as a biomarker or therapeutic target. The study of PRAMEF10 frequently involves antibody-based detection methods to characterize its expression, localization, and interactions with other cellular components.
What are the typical applications for biotin-conjugated PRAMEF10 antibodies?
Biotin-conjugated PRAMEF10 antibodies are versatile tools employed across multiple research applications. In Western blotting, these antibodies enable protein detection when paired with streptavidin-conjugated reporter systems . For ELISA assays, the biotin-conjugated antibody typically serves as the detection antibody in sandwich ELISA formats, where it binds to the captured PRAMEF10 protein and is subsequently detected using streptavidin-HRP systems . The antibody's specificity for the C-terminal region (often AA 396-424) of human PRAMEF10 makes it suitable for detecting this protein in various sample types including tissue homogenates and body fluids . The biotin conjugation provides significant advantages for signal amplification through the high-affinity biotin-streptavidin interaction, enhancing detection sensitivity in applications where protein expression might be relatively low.
How does the specificity of commercial PRAMEF10 antibodies compare across different epitope regions?
Commercial PRAMEF10 antibodies typically target specific epitope regions of the protein, with many focusing on the C-terminal region (AA 396-424) . These antibodies are generally produced by immunizing rabbits with KLH-conjugated synthetic peptides corresponding to the target epitope region . The specificity is confirmed through affinity purification and validation in applications such as Western blotting and ELISA. The epitope selection influences antibody performance across different applications - the C-terminal region (AA 396-424) is often accessible in both native and denatured protein conformations, making antibodies targeting this region versatile for multiple experimental techniques . Some manufacturers also offer antibodies targeting the middle region of PRAMEF10, which may provide alternative detection capabilities depending on protein folding and accessibility in experimental conditions . Researchers should consider that antibody reactivity is typically validated specifically for human samples, though cross-reactivity with other species may occur depending on sequence conservation.
What advantages does a biotin-conjugated PRAMEF10 antibody offer compared to other conjugates?
Biotin-conjugated PRAMEF10 antibodies offer several distinct advantages over directly labeled conjugates like PE, FITC, APC, or HRP . The primary benefit is the signal amplification potential - each biotin molecule can interact with multiple streptavidin proteins, each potentially carrying multiple reporter molecules, creating a natural signal enhancement system. This amplification is particularly valuable when detecting proteins expressed at low levels. Additionally, biotin-conjugated antibodies provide experimental flexibility, allowing researchers to select different streptavidin-reporter systems (fluorescent, enzymatic, etc.) for different applications without changing the primary antibody . The biotin-streptavidin system also demonstrates exceptional affinity (Kd ≈ 10^-15 M), contributing to stable and specific detection. In sandwich ELISA applications, biotin-conjugated antibodies serve as effective detection antibodies that can be paired with streptavidin-HRP for sensitive colorimetric readouts . These advantages make biotin-conjugated PRAMEF10 antibodies particularly valuable for researchers who require adaptability across different detection platforms or enhanced sensitivity.
What experimental techniques are most compatible with biotin-conjugated PRAMEF10 antibodies?
Biotin-conjugated PRAMEF10 antibodies demonstrate excellent compatibility with several experimental techniques. For sandwich ELISA applications, these antibodies function as detection antibodies that bind to captured PRAMEF10 and are subsequently detected using streptavidin-conjugated enzymes like HRP . In Western blotting, they can be used with streptavidin-HRP for sensitive chemiluminescent detection of PRAMEF10 protein . The antibodies are typically validated for these specific applications, with manufacturers indicating their suitability for Western blotting and ELISA methodologies . Other potentially compatible techniques include immunoprecipitation using streptavidin-coated beads, flow cytometry when paired with fluorophore-conjugated streptavidin, and immunohistochemistry or immunofluorescence when used with appropriate streptavidin-reporter systems. The versatility of the biotin-streptavidin system makes these antibodies adaptable to various research protocols where specific detection of PRAMEF10 is required.
How can I optimize ELISA protocols when using biotin-conjugated PRAMEF10 antibodies?
Optimizing ELISA protocols with biotin-conjugated PRAMEF10 antibodies requires careful attention to several key parameters to maximize sensitivity while minimizing background. The sandwich ELISA format described in the literature typically employs a capture antibody specific for PRAMEF10, followed by sample addition, then detection with the biotin-conjugated PRAMEF10 antibody, and finally signal development with streptavidin-HRP . For optimal results, researchers should conduct titration experiments to determine the ideal concentration of biotin-conjugated antibody, typically testing a range from 0.1-2 μg/ml. Blocking buffers should be carefully selected to minimize background while not interfering with the biotin-streptavidin interaction; specialized biotin-free blockers may provide superior results when using this detection system . Incubation times require optimization, with longer incubations potentially increasing sensitivity but also potentially raising background signals. For washing steps, using PBS-T (PBS with 0.05% Tween-20) and performing at least 4-5 washes between each step can significantly reduce background. When preparing standard curves, use recombinant PRAMEF10 protein diluted in the same matrix as your samples to account for matrix effects. The addition of carrier proteins (0.1-0.5% BSA) to dilution buffers can prevent non-specific antibody adsorption to plate surfaces and improve reproducibility.
What controls should be implemented when validating biotin-conjugated PRAMEF10 antibody specificity?
A comprehensive validation strategy for biotin-conjugated PRAMEF10 antibodies should include multiple controls to confirm specificity. Primary negative controls should include samples known not to express PRAMEF10 and technical controls where the primary antibody is omitted but streptavidin-reporter is still applied to assess non-specific binding . Positive controls should include samples with confirmed PRAMEF10 expression or recombinant PRAMEF10 protein. A peptide competition assay provides strong evidence of specificity - pre-incubating the antibody with excess immunizing peptide (the KLH-conjugated synthetic peptide from AA 396-424) should block binding sites and eliminate specific signal while non-specific binding would remain unaffected . For Western blot applications, molecular weight verification is essential; the detected band should appear at the expected molecular weight for PRAMEF10, and unexpected bands may indicate cross-reactivity with other proteins. Genetic manipulation controls, such as PRAMEF10 overexpression or knockdown experiments, provide functional validation of antibody specificity by demonstrating corresponding changes in signal intensity. Validation across multiple applications (e.g., Western blot, ELISA, immunofluorescence) can provide additional confidence in antibody specificity. Finally, cross-validation with antibodies targeting different PRAMEF10 epitopes can help confirm that the detected signal truly represents PRAMEF10 rather than cross-reactive proteins.
How do different epitope regions affect PRAMEF10 antibody performance across applications?
The epitope region targeted by PRAMEF10 antibodies significantly influences their performance across different experimental applications. Antibodies targeting the C-terminal region (AA 396-424), as described in the literature, offer specific performance characteristics . This region is often more accessible in both native and denatured protein conformations, making these antibodies versatile for multiple applications. In Western blotting under denaturing conditions, these C-terminal epitopes typically maintain accessibility, supporting reliable detection . For ELISA applications, the C-terminal epitope region provides consistent target recognition, particularly in sandwich ELISA formats where a different antibody serves as the capture antibody . The structural accessibility of the epitope impacts performance in native-condition applications like immunoprecipitation or flow cytometry - terminal regions are generally more exposed in folded proteins compared to internal sequences. Additionally, the specificity of the epitope region for PRAMEF10 versus other PRAME family members influences cross-reactivity potential. The C-terminal region (AA 396-424) appears to offer good specificity for PRAMEF10, though researchers should still validate this in their specific experimental systems . For applications requiring native protein detection, consideration of potential post-translational modifications near the epitope region is essential, as these could mask antibody binding sites in certain experimental conditions.
What are the structural considerations when using single-chain variable fragments (scFv) versus full antibodies for PRAMEF10 detection?
How can surface plasmon resonance (SPR) be used to characterize PRAMEF10 antibody binding kinetics?
Surface Plasmon Resonance (SPR) provides a powerful platform for real-time, label-free analysis of PRAMEF10 antibody binding kinetics. Based on published methodologies, a typical SPR experiment for characterizing PRAMEF10 antibodies would involve immobilizing streptavidin on a sensor chip (such as sensor chip CAP), followed by capturing biotinylated PRAMEF10 protein at densities of 100-500 Response Units . The PRAMEF10 antibody would then be injected over the immobilized antigen at multiple concentrations in a suitable running buffer such as HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% Surfactant P20) . A typical flow rate of 30 μL/min allows for efficient mass transport while the association phase (typically 2-5 minutes) and dissociation phase (5-15 minutes) provide data for kinetic analysis . Surface regeneration with 10 mM glycine (pH a1.5) between analyte injections enables multiple measurements on the same surface . Data analysis involves fitting the sensorgrams to appropriate binding models (1:1 Langmuir, heterogeneous ligand, or bivalent analyte models) to extract key parameters including association rate constant (ka), dissociation rate constant (kd), and equilibrium dissociation constant (KD = kd/ka). For PRAMEF10 antibodies, high-affinity interactions would typically show KD values in the nanomolar to picomolar range (10^-9-10^-11 M) . This SPR approach enables comparative analyses between different antibody formats (full antibody, Fab, scFv) and can assess how modifications like biotin conjugation might affect binding characteristics.
What approaches can I use to troubleshoot inconsistent Western blot results with biotin-conjugated PRAMEF10 antibodies?
Troubleshooting inconsistent Western blot results with biotin-conjugated PRAMEF10 antibodies requires systematic analysis of multiple experimental parameters. For sample preparation, ensure consistent protein extraction methods and include protease inhibitors to prevent PRAMEF10 degradation. When using biotin-conjugated antibodies, the blocking strategy becomes critical - standard milk-based blockers can contain endogenous biotin that interferes with the detection system; specialized biotin-free blockers or BSA-based alternatives often yield better results . The primary antibody dilution should be optimized through titration experiments, typically testing ranges from 1:500 to 1:5000 for commercial antibodies . For the detection strategy with biotin-conjugated antibodies, optimize streptavidin-HRP concentration and incubation time - excessive concentration can increase background while insufficient amounts reduce sensitivity. The washing protocol is particularly important; use TBS-T or PBS-T with at least 4-5 thorough washes after both antibody and streptavidin-HRP incubations to minimize background. If endogenous biotinylated proteins in your samples create background issues, consider pre-blocking with free streptavidin before adding the biotin-conjugated antibody. Temperature control during incubations affects reaction kinetics; maintain consistent temperatures (typically room temperature or 4°C) across experiments. For exposure settings during imaging, establish standardized exposure times based on optimal signal-to-noise ratio from control samples. Finally, always include positive controls (samples known to express PRAMEF10) and negative controls (antibody omitted) to validate results in each experiment.
How can I design validation experiments to confirm PRAMEF10 antibody specificity across different sample types?
Designing comprehensive validation experiments for PRAMEF10 antibodies requires a multi-faceted approach across different sample types. Begin with expression profiling validation by testing the antibody against a panel of cell lines or tissues with known PRAMEF10 expression levels, confirming that signal strength correlates with expected expression patterns . For molecular validation, conduct Western blotting to verify that the detected protein appears at the expected molecular weight for PRAMEF10 (~50 kDa, though this may vary with post-translational modifications) . Genetic manipulation provides powerful functional validation - use CRISPR/Cas9 to generate PRAMEF10 knockout cells, siRNA for knockdown experiments, or overexpression constructs, and confirm that antibody signal changes accordingly. Cross-reactivity assessment should evaluate potential detection of other PRAME family members by testing against recombinant proteins or cells selectively expressing individual family members. For epitope-specific validation, perform peptide competition assays using the immunizing peptide (such as the synthetic peptide from AA 396-424) to confirm that pre-incubation eliminates specific binding . When working with tissue samples, include isotype control antibodies matched to your PRAMEF10 antibody to assess non-specific binding in the specific tissue context. For ELISA applications, perform spike-and-recovery experiments across different sample matrices (serum, cell lysates, tissue homogenates) to confirm consistent detection regardless of sample composition . Finally, cross-platform validation comparing results across different techniques (Western blot, ELISA, immunohistochemistry) using the same samples provides strong evidence of specific detection across experimental contexts.