PRG2, also known as Bone Marrow Proteoglycan or Eosinophil Major Basic Protein, is a 25 kDa protein encoded by the PRG2 gene (Gene ID: 5553; UniProt ID: P13727) . It is highly expressed in eosinophil granules and placental tissues, playing roles in antiparasitic defense, immune hypersensitivity, and tissue remodeling . The mature form (206 amino acids) is derived from a 222-residue preproprotein and exhibits cytotoxic properties against parasites and mammalian cells .
This antibody is typically produced in rabbits using synthetic peptides or recombinant protein fragments from human PRG2. Key specifications include:
Western Blotting (WB): Detects PRG2 at 1:300–5,000 dilution .
Immunohistochemistry (IHC): Validated in paraffin-embedded tissues (1:50–1:500 dilution) .
Immunofluorescence (IF): Used in cell studies to localize PRG2 .
Role in Cancer: PRG2 expression correlates with tumor progression. For example, in breast cancer, PRG2 was detected in malignant tissues using IHC .
PTEN Regulation: PRG2 interacts with PTEN (phosphatase and tensin homolog), inhibiting its activity by ~50% in glioblastoma cells, which modulates PI3K/AKT signaling .
Eosinophil Function: PRG2’s cytotoxic properties contribute to eosinophil-mediated immune responses, validated via WB and IF in cellular models .
Cross-Reactivity: Predicted to react exclusively with human PRG2; no cross-reactivity with mouse or rat homologs is reported .
Sensitivity: Biotin conjugation enhances detection limits in streptavidin-HRP systems, as demonstrated in EphA2-targeting studies .
Validation: Antibody specificity is confirmed via knockdown experiments and peptide blocking .
In U87MG glioblastoma cells, PRG2 overexpression reduced PTEN-mediated hydrolysis of PIP3, restoring PI3K/AKT signaling and membrane protrusions. This was validated using WB (1:1,000 dilution) and IF, demonstrating the antibody’s utility in mechanistic studies .
PRG2 antibodies are pivotal for studying eosinophil-related diseases (e.g., asthma) and cancer. Emerging applications include biomarker discovery and therapeutic targeting, supported by biotin-streptavidin platforms for drug delivery .
PRG2, also known as Bone Marrow Proteoglycan, is a protein that functions as a cytotoxin and helminthotoxin in the immune system. It plays a significant role in inducing non-cytolytic histamine release from human basophils and is involved in antiparasitic defense mechanisms and immune hypersensitivity reactions. Importantly, the proform of PRG2 acts as a proteinase inhibitor, reducing the activity of PAPPA (Pregnancy-Associated Plasma Protein A) . The protein has been identified in various immunological contexts, with expression patterns that change under different pathological conditions, making it an important marker for immune research .
PRG2 antibodies typically demonstrate reactivity with human samples, with some variants also recognizing mouse tissues . Certain commercially available antibodies show broader reactivity profiles, including human, rat, and guinea pig samples . When selecting a PRG2 antibody for your research, it's essential to verify the specific reactivity profile of your chosen antibody, as this can significantly impact experimental outcomes in cross-species studies or when working with animal models.
Biotin conjugation provides several methodological advantages for PRG2 antibody applications. The strong affinity between biotin and streptavidin/avidin allows for enhanced sensitivity in detection systems, particularly valuable in techniques like ELISA and immunohistochemistry. This conjugation creates a versatile tool that can be integrated into various detection systems, including those utilizing streptavidin-HRP or streptavidin-fluorophore conjugates . The biotin-streptavidin binding is one of the strongest non-covalent interactions in nature, making biotin-conjugated antibodies particularly useful for detecting low-abundance proteins like PRG2 in complex biological samples.
Accurate assessment of PRG2 subcellular localization requires a methodological approach combining proper fixation, permeabilization, and detection protocols. Research has shown that PRG2 exhibits distinct localization patterns in different tissue contexts - for example, it localizes to both trophoblasts and chorionic mesoderm in some conditions while being widely expressed throughout membrane layers in others . For optimal subcellular localization studies:
Use paraformaldehyde fixation (4%) for 15-20 minutes at room temperature
Permeabilize with 0.1-0.3% Triton X-100 for 10 minutes
Block with 5% normal serum from the same species as the secondary antibody
Incubate with biotin-conjugated PRG2 antibody at optimized dilution (typically 1:100-1:500)
Detect using fluorophore-conjugated streptavidin
Co-stain with organelle markers (e.g., DAPI for nucleus, CK7 for trophoblasts)
Confocal microscopy with z-stack analysis provides the most accurate assessment of PRG2's subcellular distribution, allowing researchers to distinguish between membrane-associated, cytoplasmic, and nuclear pools of the protein .
Resolving contradictions between RNA and protein expression data for PRG2 requires a multi-faceted approach. Research has demonstrated that PRG2 typically shows a strong correlation between mRNA and protein levels (R² = 0.73, p-value = 0.031) , which is higher than many proteins that show only moderate correlation with their transcripts. When facing contradictory data:
Verify antibody specificity through western blotting and knockout/knockdown controls
Use multiple antibodies targeting different epitopes of PRG2
Perform quantitative analysis using standardized immunofluorescence protocols:
Use the minimum antibody concentration that allows detection of the lowest expression levels
Ensure measurements remain within the linear range of detection
Normalize to appropriate housekeeping proteins
Consider post-transcriptional regulation mechanisms that might affect PRG2 expression
Analyze temporal dynamics, as mRNA and protein expression may peak at different timepoints
When properly designed, studies of PRG2 should show concordance between RNA-seq and protein data, with any discrepancies potentially revealing important regulatory mechanisms .
Validating biotin-conjugated PRG2 antibodies for specificity requires a comprehensive approach:
Western blot analysis: Verify a single band of appropriate molecular weight (the expected molecular weight of PRG2 is approximately 25-28 kDa)
Peptide competition assays: Pre-incubate the antibody with synthetic peptide corresponding to the immunogen to confirm specific binding is blocked
Knockout/knockdown validation: Test the antibody in samples with genetic depletion of PRG2
Cross-reactivity assessment: Test against related proteins, particularly other proteoglycans
Tissue panel validation: Compare staining patterns across tissues with known PRG2 expression profiles
Antibody dilution series: Perform titration experiments to determine optimal concentration that maximizes signal-to-noise ratio
Multiple antibody comparison: Use antibodies targeting different epitopes of PRG2 (e.g., internal region vs. amino acids 51-150) to confirm consistent staining patterns
The most rigorous validation combines multiple approaches and includes appropriate negative controls for each experiment.
The optimal ELISA protocol for biotin-conjugated PRG2 antibodies involves:
Plate preparation:
Coat high-binding 96-well plates with capture antibody (anti-PRG2, unconjugated) at 1-2 μg/ml in coating buffer (carbonate-bicarbonate, pH 9.6)
Incubate overnight at 4°C
Wash 3-5 times with wash buffer (PBS with 0.05% Tween-20)
Blocking and sample addition:
Block with 1-5% BSA in PBS for 1-2 hours at room temperature
Add samples and standards diluted in sample diluent
Incubate for 2 hours at room temperature or overnight at 4°C
Detection:
Add biotin-conjugated PRG2 detection antibody (typically diluted 1:100 in antibody dilution buffer)
Incubate for 1-2 hours at room temperature
Wash 3-5 times with wash buffer
Add HRP-streptavidin conjugate (SABC) working solution (diluted 1:100 in SABC dilution buffer)
Incubate for 30-60 minutes at 37°C
Wash 5 times
Development and analysis:
This protocol typically provides a sensitive detection range of 15.6-1000 pg/ml for PRG2 .
Optimizing biotin-conjugated PRG2 antibodies for immunohistochemistry requires careful attention to several methodological factors:
Tissue preparation and antigen retrieval:
Formalin-fixed paraffin-embedded (FFPE) sections should be deparaffinized and rehydrated
Heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) for 20 minutes
Allow sections to cool to room temperature for 20 minutes
Blocking endogenous biotin and peroxidase:
Block endogenous peroxidase with 0.3% H₂O₂ in methanol for 10 minutes
Critical step: Use a commercial biotin blocking system to block endogenous biotin
Block non-specific binding with 5-10% normal serum from the same species as the secondary antibody
Antibody dilution optimization:
Detection system:
Apply HRP-streptavidin conjugate at 1:100-1:500 dilution
Develop with DAB or other appropriate chromogen
Counterstain with hematoxylin for nuclear visualization
This protocol has been validated for detecting PRG2 in placental tissues, with specific localization to trophoblasts and chorionic mesoderm under normal conditions .
High background is a common challenge when working with biotin-conjugated antibodies. Effective approaches to overcome this issue include:
Endogenous biotin blocking:
Use commercial avidin/biotin blocking kits
Alternative method: Incubate sections with unconjugated avidin (10-20 μg/ml) followed by biotin (50 μg/ml)
Buffer optimization:
Add 0.1-0.5% Tween-20 to wash buffers
Include 0.1-0.3% Triton X-100 in antibody diluent for better penetration
Add 0.1-0.5% BSA to antibody diluent to reduce non-specific binding
Antibody optimization strategies:
Further dilute the biotin-conjugated PRG2 antibody
Reduce incubation time from overnight to 1-2 hours at room temperature
Pre-absorb antibody with tissue homogenate from species of interest
Sample-specific considerations:
For tissues rich in endogenous biotin (liver, kidney, brain):
Use alternative detection methods when possible
Apply more stringent biotin blocking protocols
For tissues with high background:
Increase washing steps (5-7 washes of 5 minutes each)
Use 0.3M NaCl in wash buffer to reduce ionic interactions
Controls for troubleshooting:
Omit primary antibody to assess secondary antibody/detection system background
Use isotype control to assess non-specific binding
Include tissue known to be negative for PRG2 as a negative control
These approaches have been validated in studies examining PRG2 expression in various tissue samples, including placental tissues where precise localization is critical for accurate interpretation .
Effective multiplex immunofluorescence with biotin-conjugated PRG2 antibodies requires careful planning and execution:
Panel design considerations:
Sequential staining protocol:
Apply unconjugated primary antibodies simultaneously or sequentially
Apply species-specific secondary antibodies (ensure no cross-reactivity)
Apply biotin-conjugated PRG2 antibody
Detect with fluorophore-conjugated streptavidin (e.g., streptavidin-Alexa Fluor 647)
Signal amplification options:
For weak PRG2 signals, implement tyramide signal amplification (TSA)
Use streptavidin-HRP followed by tyramide-fluorophore conjugates
Include HRP quenching steps between rounds if detecting multiple targets
Quality control measures:
Include single-stained controls for each antibody
Perform fluorescence minus one (FMO) controls
Use spectral unmixing if available on imaging platform
Quantitative analysis approach:
Capture images at identical exposure settings
Measure fluorescence intensity in defined cellular compartments
Normalize to appropriate reference markers
Calculate co-localization coefficients if assessing protein interactions
This approach has been validated in research examining PRG2 expression in relation to other markers in placental tissues, demonstrating the value of multiplex analysis for understanding complex expression patterns .
When utilizing biotin-conjugated PRG2 antibodies for flow cytometry, several methodological considerations are critical:
Sample preparation:
For cell suspensions: Fix with 2-4% paraformaldehyde for 10-15 minutes
For intracellular staining: Permeabilize with 0.1% saponin or 0.3% Triton X-100
Maintain cold chain (4°C) for all steps prior to fixation
Staining protocol optimization:
Titrate antibody to determine optimal concentration (typically 0.1-1 μg per million cells)
Use a two-step approach: biotin-conjugated PRG2 antibody followed by fluorophore-conjugated streptavidin
Include viability dye to exclude dead cells
Block Fc receptors with 5% normal serum or commercial Fc block
Controls required:
Unstained cells
Single-color controls for compensation
Fluorescence minus one (FMO) control
Isotype control conjugated to biotin
Biological negative control (cells known not to express PRG2)
Analysis strategies:
Gate on single, viable cells
Compare PRG2 expression levels across different cell populations
Consider co-expression with lineage markers
Analyze both percentage positive and mean fluorescence intensity
Special considerations for PRG2:
As PRG2 can be secreted, intracellular staining protocols are often necessary
For certain cell types, protein transport inhibitors (like Brefeldin A) may improve detection
In some contexts, PRG2 may be bound to cell surface via interactions with other molecules
Flow cytometry is particularly valuable for quantifying PRG2 expression across different cell populations and can complement tissue-based analyses that provide spatial information.
Proper normalization of quantitative PRG2 data is essential for meaningful comparisons across experiments:
Western blotting normalization approaches:
Normalize to total protein (measured by Ponceau S or REVERT total protein stain)
Alternatively, normalize to housekeeping proteins (GAPDH, β-actin, tubulin)
Calculate relative expression as PRG2/reference protein
Present data as fold change relative to control samples
Immunofluorescence quantification:
Measure fluorescence intensity within defined regions of interest
Subtract background signal from adjacent negative areas
When comparing across tissues or experiments:
ELISA data analysis:
Generate standard curve using known concentrations
Use four-parameter logistic regression for curve fitting
Ensure all sample measurements fall within the linear range of the standard curve
Express results as absolute concentration (pg/ml) based on standard curve
Statistical considerations:
Research has demonstrated that proper normalization is critical for accurate interpretation of PRG2 expression data, particularly when comparing across different pathological conditions .
Interpretation of PRG2 expression data presents several common pitfalls that researchers should be aware of:
By addressing these common pitfalls, researchers can ensure more robust and reliable interpretation of PRG2 expression data across different experimental contexts.
Biotin-conjugated PRG2 antibodies have emerged as valuable tools in advancing our understanding of placental pathologies through several methodological approaches:
Molecular signature development:
PRG2 has been identified as a key component of molecular signatures in placental disorders
Research demonstrates PRG2 is significantly upregulated in both placenta previa and placenta accreta spectrum (PAS) disorders
Quantitative immunofluorescence using biotin-conjugated antibodies has confirmed these findings across multiple patient cohorts
Cellular localization insights:
High-resolution imaging with biotin-conjugated antibodies has revealed that PRG2 shows distinct localization patterns in different placental pathologies
In normal placentas, PRG2 localizes primarily to trophoblasts and chorionic mesoderm
In previa and percreta samples, PRG2 expression extends throughout membrane layers (except amnion)
These patterns suggest altered cellular programming in pathological states
Correlation with clinical outcomes:
Studies correlating PRG2 expression levels with clinical parameters are providing insights into potential prognostic applications
The strong correlation between PRG2 mRNA and protein levels (R² = 0.73, p-value = 0.031) supports its reliability as a biomarker
This correlation exceeds typical protein-mRNA relationships, which often show only moderate correlation (R² ≈ 0.4)
Multi-marker analysis approaches:
Combined analysis of PRG2 with other markers (such as AQPEP and HLA-G) is revealing complex molecular signatures
These signatures may have diagnostic and prognostic value in managing placental disorders
These advances suggest promising future directions for biotin-conjugated PRG2 antibodies in both research and clinical applications related to placental pathologies.
Several emerging techniques are significantly enhancing the utility of biotin-conjugated antibodies in PRG2 research:
Spatial transcriptomics integration:
Combining biotin-conjugated PRG2 antibody staining with spatial transcriptomics
This allows correlation of protein localization with gene expression profiles at single-cell resolution
Particularly valuable for understanding PRG2's role in complex tissues like placenta
Proximity ligation assays (PLA):
Using biotin-conjugated PRG2 antibodies in combination with antibodies against potential interaction partners
PLA provides visualization of protein-protein interactions in situ
Can reveal functional relationships between PRG2 and other proteins in the cellular context
Mass cytometry (CyTOF) applications:
Metal-tagged streptavidin can be used with biotin-conjugated PRG2 antibodies
Allows simultaneous detection of dozens of markers at single-cell resolution
Particularly valuable for comprehensive immune cell profiling in inflammatory conditions
Organoid-based validation systems:
Using biotin-conjugated PRG2 antibodies to validate expression in 3D organoid cultures
Provides more physiologically relevant models than traditional 2D cultures
Allows for manipulation of PRG2 expression in controlled environments
Super-resolution microscopy techniques:
Techniques like STORM and PALM provide nanoscale resolution of PRG2 localization
Biotin-streptavidin pairs are ideal for these applications due to high specificity
Reveals subcellular distribution patterns not visible with conventional microscopy