PRKACA biotin-conjugated antibodies are polyclonal or monoclonal antibodies chemically linked to biotin, targeting the PRKACA protein. PRKACA is a catalytic subunit of cAMP-dependent protein kinase A, involved in critical cellular processes like metabolism, gene expression, and signal transduction . Biotinylation enhances detection sensitivity in techniques such as ELISA, Western blotting (WB), and immunohistochemistry (IHC) .
ELISA: Used for quantitative detection of PRKACA in serum or cell lysates .
Western Blot: Detects PRKACA at ~38–43 kDa in human, mouse, and rat samples .
Immunohistochemistry: Localizes PRKACA in tissues like breast cancer and testis .
PRKACA antibodies aid in studying:
Cancer Research: Overexpression in fibrolamellar hepatocellular carcinoma (FL-HCC) linked to the DNAJB1-PRKACA fusion driver .
Metabolic Regulation: Impact on glucose homeostasis and adipogenesis .
Specificity: Targets unique epitopes within PRKACA (e.g., 233–351AA or 495–659AA) .
Sensitivity: Biotin conjugation reduces background noise, improving signal-to-noise ratios in complex samples .
Cross-Reactivity: Validated in human, mouse, and rat models .
PRKACA antibodies have been critical in identifying the DNAJB1-PRKACA fusion protein in FL-HCC, a rare liver cancer. This fusion generates unique neoepitopes targeted by immunotherapies .
PRKACA (protein kinase, cAMP-dependent, catalytic, alpha) is a key catalytic subunit of protein kinase A with a molecular weight of approximately 40.6 kDa (calculated) and 38-43 kDa (observed in experiments). This protein is widely expressed across multiple tissue types and functions as a member of the AGC Ser/Thr protein kinase family . PRKACA plays crucial roles in GPCR signaling pathways and carbohydrate metabolism and homeostasis, making it a significant target in various research fields .
The protein localizes to multiple cellular compartments including the cell membrane, nucleus, cytoplasmic vesicles, mitochondria, and cytoplasm . Its involvement in numerous signaling pathways makes it relevant to research in oncology, metabolism, and cell signaling. Particularly notable is the DNAJB1-PRKACA fusion protein, which serves as an oncogenic driver in fibrolamellar hepatocellular carcinoma, a lethal disease with limited therapeutic options .
PRKACA antibodies have demonstrated reactivity with samples from multiple species including human, mouse, and rat tissues and cell lines . The following table summarizes optimal sample types based on validated experimental data:
| Application | Validated Sample Types | Recommended Dilutions |
|---|---|---|
| Western Blot (WB) | HeLa cells, Neuro-2a cells, SH-SY5Y cells, mouse/rat testis tissue | 1:500-1:6000 |
| Immunohistochemistry (IHC) | Human breast cancer tissue, human testis tissue, human heart tissue | 1:50-1:500 |
| Immunoprecipitation (IP) | HeLa cells | 0.5-4.0 μg per 1-3 mg of total protein lysate |
| Immunofluorescence (IF) | U2OS cells | 1:50-1:500 |
For IHC applications, antigen retrieval can be performed using TE buffer at pH 9.0 or alternatively with citrate buffer at pH 6.0 . The high conservation of PRKACA across species enables cross-reactivity, providing flexibility in experimental design across different model systems.
Validating antibody specificity is essential for reliable experimental outcomes. For PRKACA antibodies, consider the following validation approaches:
Positive and negative controls: Use cell lines with known PRKACA expression levels. Based on published data, HeLa, Neuro-2a, and SH-SY5Y cells serve as reliable positive controls .
Molecular weight verification: Confirm that the detected band falls within the expected range of 38-43 kDa on Western blots .
Knockdown/knockout validation: Verify specificity by comparing detection between wildtype and PRKACA-depleted samples. Published literature includes knockout validation for certain PRKACA antibodies .
Cross-verification: Compare results using multiple antibodies targeting different epitopes of PRKACA to ensure consistent detection patterns.
Blocking peptide competition: Pre-incubate the antibody with the immunogen peptide to confirm specificity through signal reduction.
The antibody's specificity can be further confirmed by comparing experimental results with published data where PRKACA has been detected in similar experimental conditions.
When incorporating PRKACA antibodies into multiplex immunoassays, several technical considerations should be addressed:
Antibody compatibility: Select antibodies raised in different host species to avoid cross-reactivity in multiple labeling experiments. PRKACA antibodies are available in rabbit host format, which should be paired with antibodies from other species .
Signal optimization: For fluorescence-based detection methods, consider the following parameters:
Fixation: 4% paraformaldehyde for 15-20 minutes at room temperature
Permeabilization: 0.1-0.3% Triton X-100 for 5-10 minutes
Blocking: 5% normal serum (matched to secondary antibody host) with 1% BSA for 30-60 minutes
Detection strategy: For biotin-conjugated antibodies, use streptavidin coupled to a fluorophore with minimal spectral overlap with other detection reagents. Optimal dilution ranges for biotin-conjugated antibodies typically require titration starting at 1:200-1:500.
Controls: Include single-stained controls to assess bleed-through and secondary-only controls to evaluate background signal.
Sequential staining: Consider sequential rather than simultaneous staining protocols when using antibodies with similar properties to minimize cross-reactivity.
When optimizing multiplex protocols, each antibody should be individually validated before combining into the multiplex system to ensure specificity is maintained in the more complex environment.
Inconsistent antibody performance can arise from multiple factors. The following troubleshooting guide addresses common issues specifically relevant to PRKACA antibody applications:
Sample preparation variations:
Epitope accessibility issues:
PRKACA has multiple subcellular localizations (membrane, nucleus, cytoplasm, mitochondria)
Different fixation and permeabilization protocols may be required depending on the targeted subcellular compartment
For membrane-associated PRKACA, mild detergents like 0.1% Tween-20 may be preferable to stronger agents
Antibody dilution optimization:
Post-translational modifications:
PRKACA function is regulated by phosphorylation
Phosphorylation-dependent epitope masking may affect antibody recognition
Consider phosphatase treatment of samples if epitope recognition is influenced by phosphorylation status
Lot-to-lot variations:
Document lot numbers and maintain reference samples for comparison when switching antibody lots
Request certificate of analysis with batch-specific validation data from suppliers
For biotin-conjugated antibodies specifically, additional considerations include streptavidin blocking steps for endogenous biotin and optimizing streptavidin-conjugate concentrations to minimize background while maintaining sensitivity.
The DNAJB1-PRKACA fusion protein represents a unique oncogenic driver in fibrolamellar hepatocellular carcinoma (FL-HCC) . When studying this fusion protein, researchers should consider:
Antibody epitope location:
Select antibodies targeting the PRKACA portion that is preserved in the fusion protein
Differential detection between wild-type PRKACA and the fusion protein may require antibodies targeting the fusion junction
Expression system considerations:
The fusion protein maintains kinase activity but with altered substrate specificity
Expression levels may differ from wild-type PRKACA
Control experiments should include both wild-type PRKACA and fusion protein-expressing systems
Immunological significance:
Technical validation approaches:
The DNAJB1-PRKACA fusion has been successfully targeted with peptide-based immunotherapy, highlighting its importance as both a diagnostic marker and therapeutic target in FL-HCC research .
Biotin conjugation provides specific advantages and requires methodological adjustments when working with PRKACA antibodies:
Signal amplification considerations:
Biotin-streptavidin interaction provides strong (Kd ~10^-15 M) and stable binding
Multiple biotin molecules per antibody can increase detection sensitivity
Optimal working dilutions for biotinylated antibodies may differ from unconjugated versions, typically requiring 2-3 fold higher dilutions
Endogenous biotin interference:
Certain tissues (liver, kidney, brain) contain high levels of endogenous biotin
Incorporate avidin/streptavidin blocking steps (10-15 minutes with commercial blocking kit) before applying biotinylated antibodies
Consider the use of specialized blocking reagents (e.g., streptavidin/biotin blocking kit) for tissues with high endogenous biotin
Detection system selection:
Enzymatic detection: Streptavidin-HRP or streptavidin-AP followed by appropriate substrate
Fluorescent detection: Streptavidin conjugated to fluorophores (Alexa 488, 555, 647)
Choosing detection systems with minimal background in the tissue/cells of interest
Storage considerations:
Biotin-conjugated antibodies typically maintain stability at -20°C with 50% glycerol
Avoid repeated freeze-thaw cycles which can degrade both the antibody and biotin moiety
Prepare single-use aliquots when possible
Multiplexing advantages:
Biotin-streptavidin pairing enables flexible secondary detection systems
Compatible with TSA (tyramide signal amplification) for ultra-sensitive detection
Can be combined with directly labeled antibodies in multiplex experiments
When directly comparing results between biotinylated and non-biotinylated PRKACA antibodies, include appropriate controls to account for potential differences in sensitivity and background.
Detecting PRKACA in challenging samples requires optimized protocols tailored to specific sample characteristics:
Formalin-fixed paraffin-embedded (FFPE) tissues:
Extended antigen retrieval: 20-30 minutes in TE buffer pH 9.0 at 95-100°C
Signal amplification: Consider tyramide signal amplification for low-abundance detection
Background reduction: Include additional blocking steps (0.3% hydrogen peroxide, avidin/biotin blocking)
Optimal dilution range: 1:50-1:200 (more concentrated than fresh samples)
Brain tissues with high lipid content:
Extended fixation: 24-48 hours in 4% PFA
Permeabilization enhancement: Add 0.1-0.3% Triton X-100 to all antibody diluents
Background reduction: Include 0.1-0.3 M glycine to reduce autofluorescence
Consider antigen retrieval even for frozen sections to improve epitope accessibility
Archived/degraded samples:
Target selection: Focus on stable epitopes less affected by long-term storage
Signal recovery: Apply heat-induced epitope retrieval with pH optimization (test both acidic and basic buffers)
Alternative detection: Consider proximity ligation assay for improved specificity in partially degraded samples
Samples with high background:
For tissues with high endogenous peroxidase: Extended hydrogen peroxide blocking (0.3-3% for 15-30 minutes)
For highly autofluorescent tissues: Pretreatment with Sudan Black B (0.1-0.3%) or commercial autofluorescence quenchers
For tissues with high IgG content: Include mouse/rabbit IgG blocking steps before primary antibody application
Low abundance detection:
These specialized protocols should be validated on control samples before application to experimental samples of high value or limited availability.
Investigating PRKACA protein interactions and signaling requires specialized experimental approaches:
Co-immunoprecipitation protocols:
Lysis buffer optimization: Use non-denaturing buffers containing 1% NP-40 or 0.5% Triton X-100 with protease inhibitors
Antibody selection: Use antibodies validated for IP (0.5-4.0 μg per reaction)
Capture approach: Protein A/G beads or directly conjugated magnetic beads
Controls: Include IgG control immunoprecipitations and input lysate lanes
Detection: Blot for both PRKACA and suspected interaction partners
Proximity ligation assay (PLA) for in situ interaction studies:
Cell preparation: 4% PFA fixation for 15 minutes, 0.2% Triton X-100 permeabilization
Antibody selection: Combine PRKACA antibody with antibodies against suspected interaction partners (from different host species)
Detection: Follow commercial PLA protocols with optimization of antibody concentrations
Controls: Include single primary antibody controls and known interaction pairs
Functional interaction assays:
Kinase activity assays: Measure PRKACA activity using phospho-specific antibodies against known substrates
Inhibitor studies: Compare phosphorylation patterns with and without selective PKA inhibitors
Subcellular fractionation: Track compartment-specific interactions through biochemical separation followed by immunoblotting
FRET/BRET approaches: For dynamic, live-cell monitoring of protein-protein interactions
Mass spectrometry-based interactome analysis:
Immunoprecipitate PRKACA using validated antibodies (optimally 2-5 μg per IP)
Process samples for LC-MS/MS analysis
Employ appropriate controls (IgG IPs, competing peptides)
Validate key interactions through orthogonal methods (co-IP, PLA)
When studying the DNAJB1-PRKACA fusion protein specifically, consider comparative interaction analyses between wild-type PRKACA and the fusion protein to identify altered protein interactions that may contribute to oncogenic activity .
Discrepancies between protein detection and gene expression data are common in biological research. For PRKACA specifically:
Potential sources of discrepancy:
Post-transcriptional regulation: miRNA targeting of PRKACA mRNA
Post-translational modifications affecting epitope recognition
Protein stability differences across tissue types
Subcellular localization changes affecting extraction efficiency
Presence of multiple isoforms (up to 2 different isoforms have been reported)
Validation approaches:
Multi-antibody verification: Use antibodies targeting different PRKACA epitopes
Orthogonal methods: Combine Western blot with targeted mass spectrometry
Subcellular fractionation: Assess compartment-specific distribution
Translation inhibition studies: Evaluate protein half-life using cycloheximide chase
Data integration frameworks:
Normalize protein data to housekeeping proteins and mRNA data to reference genes
Consider time-course studies to capture potential delays between transcription and translation
Employ statistical methods designed for multi-omics data integration
Biological interpretation:
PRKACA function is heavily regulated post-translationally
Protein activity may not correlate directly with abundance
Consider activity-based assays alongside abundance measurements
For the most comprehensive understanding, researchers should combine protein detection methods with activity measurements and localization studies, particularly when transcriptomic and proteomic data appear contradictory.
PRKACA has emerging importance in immunotherapy research, particularly through the DNAJB1-PRKACA fusion in FL-HCC . When using PRKACA antibodies in this context, consider:
Epitope selection for therapeutic targeting:
Validation for immunotherapy research:
Applications in monitoring therapeutic responses:
Track changes in PRKACA or fusion protein expression during treatment
Monitor neoantigen-specific T cell responses using matched antibody-based detection systems
Assess tumor infiltrating lymphocytes in relation to PRKACA/fusion protein expression
Translational considerations:
DNAJB1-PRKACA fusion-derived peptide vaccines have shown promise in inducing multifunctional CD4+ T cells with an activated T-helper 1 phenotype
Combining antibody-based detection with T cell receptor sequencing can provide comprehensive immune response monitoring
Consider combinatorial approaches targeting both the fusion protein and the tumor microenvironment
For immunotherapy applications, the specificity of antibodies for distinguishing between wild-type PRKACA and the fusion protein is particularly critical for accurate patient stratification and response monitoring.
Emerging technologies are expanding the utility of PRKACA antibodies in several innovative directions:
Single-cell proteomics applications:
Integration with mass cytometry (CyTOF) for high-dimensional analysis
Adaptation to microfluidic-based single-cell Western blotting
Incorporation into spatial proteomics platforms for tissue-level analysis
Development of photocleavable antibody-DNA conjugates for single-cell sequencing
Live-cell imaging innovations:
Development of conformation-sensitive antibody derivatives
Integration with optogenetic systems for activity-dependent detection
Nanobody-based approaches for improved intracellular penetration
FRET-based biosensors incorporating antibody-derived binding domains
Therapeutic antibody development potential:
Engineered antibodies recognizing the DNAJB1-PRKACA fusion junction
Antibody-drug conjugates specifically targeting fusion-positive cells
Bispecific antibodies linking T cells to fusion-expressing cancer cells
CAR-T cell development based on antibody-derived binding domains
Structural biology applications:
Antibody-assisted cryo-EM for capturing specific PRKACA conformational states
Antibody-mediated crystallization of challenging protein complexes
In-cell structural studies combining antibody detection with proximity labeling
These technological innovations are poised to transform both basic research into PRKACA biology and translational applications targeting PRKACA-related pathologies, particularly in the context of the DNAJB1-PRKACA fusion protein's role in fibrolamellar hepatocellular carcinoma .
Standardization efforts are critical for enhancing reproducibility in PRKACA antibody applications:
Validation reporting standards:
Document complete validation protocols including positive and negative controls
Report antibody catalog numbers, lot numbers, and dilutions used
Include details on sample preparation, blocking, and detection methods
Share raw unprocessed images alongside processed data
Cross-laboratory validation initiatives:
Participate in multi-laboratory validation studies
Share standard operating procedures through repositories
Contribute to antibody validation databases with application-specific data
Develop reference standards for PRKACA detection across applications
New technology implementation:
Adopt orthogonal validation methods (mass spectrometry, CRISPR controls)
Implement quantitative metrics for antibody performance
Explore automation to reduce technical variability
Adopt digital pathology tools for standardized image analysis
Community resource development:
Contribute to open repositories of validated protocols
Participate in collaborative projects establishing reference standards
Share detailed methodological notes beyond standard Materials and Methods sections
Develop and share positive and negative control materials
Researchers can significantly advance the field by adhering to these standardization practices and contributing to community resources that enhance reproducibility in PRKACA antibody applications across basic research and clinical contexts.