BRD3 antibodies target the BRD3 protein (UniProt: Q15059), which contains two tandem bromodomains and an extraterminal (ET) motif. These antibodies are critical for:
Detection: Identifying BRD3 expression in tissues (e.g., colorectal cancer, synovial fibroblasts) .
Functional Studies: Elucidating BRD3's role in cell cycle regulation, inflammation, and transcriptional elongation .
Therapeutic Research: Investigating BRD3 as a potential target for cancer therapies .
Tumor Suppression: BRD3 knockdown increased colorectal cancer (CRC) cell proliferation, while overexpression inhibited tumor growth in vitro and in vivo via p21-mediated cell cycle arrest .
Nuclear TYRO3 Interaction: BRD3 binds nuclear TYRO3 in CRC, promoting oncogenic pathways like SNAI1 and CDC27 expression .
Rheumatoid Arthritis (RA): BRD3 regulates cytokine/chemokine expression (e.g., CCL2, CXCL1) in synovial fibroblasts, correlating with synovial inflammatory scores .
Chromatin Remodeling: BRD3 binds acetylated histones (H3K18ac) and non-histone proteins (e.g., GATA1), facilitating transcriptional activation .
Western Blot:
Immunohistochemistry (IHC):
Flow Cytometry:
BRD3 (Bromodomain Containing 3) is a critical chromatin reader that recognizes and binds acetylated histones, thereby controlling gene expression and remodeling chromatin structures . As a member of the BET (Bromodomain Extra Terminal) family, BRD3 contains two tandem bromodomains (BD1 and BD2) and an extraterminal (ET) domain . BRD3 recruits transcription factors and coactivators to target gene sites and activates RNA polymerase II machinery for transcriptional elongation . Its importance in epigenetic research stems from its ability to bind acetylated lysine residues on histones H2A, H2B, H3, and H4, as well as non-histone acetylated proteins like GATA1 and GATA2 . Recent studies have also implicated BRD3 in inflammatory responses, stress adaptation, and various disease mechanisms, making it a valuable target for research into transcriptional regulation and potential therapeutic interventions .
BRD3 antibodies are employed in multiple research techniques including:
Western Blotting (WB): Used for detecting and quantifying BRD3 protein expression with recommended dilutions ranging from 1:500 to 1:2000
Chromatin Immunoprecipitation (ChIP and ChIP-Seq): Critical for studying BRD3 genomic occupancy and its interactions with chromatin
Immunoprecipitation (IP): For isolating BRD3 protein complexes and studying protein-protein interactions
Immunohistochemistry (IHC): For visualizing BRD3 expression in tissue samples
Immunofluorescence/Immunocytochemistry (IF/ICC): For subcellular localization studies
Flow Cytometry: For analyzing BRD3 expression in cell populations
Most commercially available BRD3 antibodies show reactivity with human samples, with some also validated for mouse and rat models .
Selection should be based on:
Application compatibility: Verify the antibody has been validated for your specific application (WB, ChIP, IF, etc.)
Species reactivity: Ensure compatibility with your experimental model organism (human, mouse, rat)
Antibody type: Consider whether polyclonal or monoclonal is more appropriate:
Epitope location: Some antibodies target N-terminal regions while others target C-terminal regions; this matters when studying specific domains or truncated variants
Validation data: Review published literature citing the antibody and examine manufacturer validation data including positive controls
Protocol compatibility: Consider buffer compatibility and validated dilution ranges for your specific protocol
For ChIP applications specifically, seek antibodies with demonstrated ChIP-grade quality and published ChIP-seq datasets .
For successful BRD3 ChIP experiments:
Antibody amount: Typically 1-10 μL per ChIP reaction, with Cell Signaling Technology recommending 1:50 dilution for their BRD3 (E3D5N) Rabbit mAb
Cross-linking conditions: Standard 1% formaldehyde for 10 minutes at room temperature is typically sufficient for BRD3 ChIP
Sonication parameters: Aim for chromatin fragments of 200-500 bp for optimal BRD3 binding site resolution
Controls: Include:
Washing stringency: Multiple washes with increasing salt concentration to reduce background
Elution and reversal: Standard elution buffers and overnight reversal of cross-linking at 65°C
When designing primers for ChIP-qPCR validation, target regions with known histone acetylation marks, particularly H3K18ac, which has been shown to interact with BRD3 .
Based on published methodologies , a comprehensive approach includes:
Co-immunoprecipitation (Co-IP):
Use anti-BRD3 antibody to pull down protein complexes
Western blot for associated transcription factors (e.g., GATA1)
Include acetylation inhibitors as negative controls to verify acetylation-dependent interactions
Sequential ChIP (Re-ChIP):
First ChIP with anti-BRD3 antibody
Second ChIP with anti-GATA1 antibody
Analyze enrichment at common target genes
Genome-wide occupancy analysis:
Perform parallel ChIP-seq for BRD3 and transcription factors
Compare binding profiles using bioinformatic approaches
Focus on:
Co-occupied sites
Sites with differential occupancy under various conditions
Correlation with histone acetylation marks
Functional validation:
BRD3 knockdown/knockout followed by transcription factor ChIP
Analysis of target gene expression
Use of BET inhibitors to disrupt interactions
This experimental design has successfully revealed that BRD3 and GATA1 physically interact in an acetylation-dependent manner, and that BRD3 occupies most GATA1-occupied regulatory DNA elements .
A thorough validation approach should include:
Molecular controls:
Cross-reactivity assessment:
Multiple technique validation:
Epitope blocking experiments:
Pre-incubate antibody with immunizing peptide
Should eliminate specific signal in all applications
Alternative antibody comparison:
When publishing, include complete validation data and antibody catalog information to ensure reproducibility.
The calculated molecular weight of BRD3 is approximately 61 kDa (556 amino acids), yet observed molecular weights typically range from 95-105 kDa . This discrepancy may result from:
Post-translational modifications:
Phosphorylation: BRD3 contains multiple potential phosphorylation sites
Acetylation: As an acetyl-lysine binding protein, BRD3 itself may be acetylated
SUMOylation or ubiquitination: Can significantly increase apparent molecular weight
Protein isoforms:
Alternative splicing generating different BRD3 variants
Tissue or cell type-specific isoform expression
Technical factors:
Incomplete denaturation: Particularly common with chromatin-associated proteins
Running buffer composition affecting mobility
Gel percentage affecting resolution of higher molecular weight proteins
Experimental conditions:
Sample preparation method (lysis buffers, detergent concentration)
Reducing vs. non-reducing conditions
Heat denaturation time and temperature
To address these variations, include positive controls with known BRD3 expression, optimize sample preparation protocols, and compare results with multiple BRD3 antibodies targeting different epitopes .
Based on reported challenges with BRD3 ChIP signal-to-noise ratios , consider these optimization strategies:
Antibody considerations:
Test multiple BRD3 antibodies; monoclonal antibodies often provide better specificity
Titrate antibody concentration (too much can increase background)
Pre-clear chromatin with protein A/G beads before antibody addition
Cross-linking optimization:
Adjust formaldehyde concentration (typically 0.75-1.5%)
Test dual cross-linking with additional agents (DSG, EGS) for improved protein-protein cross-linking
Fine-tune cross-linking time (8-12 minutes typically optimal)
Sonication parameters:
Ensure consistent fragment size (200-500bp)
Avoid over-sonication which can damage epitopes
Verify sonication efficiency by gel electrophoresis before proceeding
Washing conditions:
Increase wash stringency with higher salt concentrations
Add detergents (0.1% SDS, 1% Triton X-100) to reduce non-specific binding
Increase number of washes while maintaining gentle agitation
Data analysis approaches:
Use appropriate peak-calling algorithms optimized for transcription factors
Include input normalization and IgG controls
Consider using spike-in controls for quantitative normalization
These optimizations address the challenges noted in the literature where "the signal-to-noise ratio of the Brd3A ChIP was not as high as that of GATA1" .
Common IHC challenges with BRD3 antibodies include:
Antigen retrieval issues:
Non-specific staining:
Fixation artifacts:
Problem: Overfixation masking epitopes or underfixation causing tissue degradation
Solution: Standardize fixation time (18-24 hours in 10% neutral buffered formalin is typically optimal)
Variable expression levels:
Cross-reactivity with other BET proteins:
Problem: Similar sequence homology between BRD2, BRD3, and BRD4
Solution: Validate antibody specificity with peptide competition assays and BRD3-knockout controls
For optimal results, follow tissue-specific recommendations for antigen retrieval and include comprehensive controls in each staining batch .
Based on genome-wide occupancy studies , BRD3 ChIP-seq data interpretation requires nuanced analysis:
The genome-wide analysis by Lamonica et al. revealed that "Brd3 is versatile in nature, occupying both transcriptionally active and inactive euchromatin" , highlighting the importance of comprehensive data integration for accurate interpretation.
To distinguish direct from indirect effects:
Temporal analysis:
Monitor gene expression changes at multiple early time points after BRD3 inhibition
Direct targets typically show rapid expression changes (within hours)
Indirect effects accumulate at later time points
Dose-response relationships:
Direct targets often show dose-dependent responses to BRD3 inhibitors
Compare with BRD3 knockdown/knockout phenotypes
Genomic occupancy correlation:
Rescue experiments:
Reintroduce wildtype BRD3 or bromodomain mutants
Direct targets should be rescued by wildtype but not binding-deficient mutants
Mechanistic validation:
For presumed direct targets, perform detailed analysis of:
Transcription factor recruitment
RNA polymerase II occupancy
Histone modification changes
Chromatin accessibility alterations
These approaches help distinguish primary transcriptional effects from secondary responses to BRD3 modulation.
When comparing results from different antibodies or systems:
Antibody characteristics:
Epitope location: Antibodies targeting different regions (N-terminal vs. C-terminal) may detect different isoforms or post-translationally modified forms
Clonality: Polyclonal antibodies detect multiple epitopes while monoclonals recognize single epitopes
Validation methods: Compare the validation techniques used for each antibody
Experimental system variables:
Cell/tissue types: BRD3 function can be context-dependent across different tissues
Species differences: Consider conservation of the BRD3 epitope across species
Expression levels: Endogenous vs. overexpression systems yield different results
Technical parameters:
Protocol differences: Variations in fixation, extraction, or detection methods
Quantification approaches: Normalization methods and reference genes/proteins
Batch effects: Account for experimental variation between batches
Data reporting standards:
Complete antibody information: Catalog numbers, lot numbers, dilutions
Detailed methods: Include all protocol parameters for reproducibility
Raw data availability: Where possible, provide access to raw data
When conflicting results are observed, consider performing side-by-side comparisons with multiple antibodies under identical conditions to identify the source of variation.
Recent research has revealed that BRD3 undergoes liquid-liquid phase separation (LLPS) upon binding to lncRNA DIGIT , presenting exciting research opportunities:
Immunofluorescence approaches:
Use high-resolution microscopy with BRD3 antibodies to visualize condensate formation
Co-staining with RNA markers to verify RNA-dependent condensates
Live-cell imaging with fluorescently tagged BRD3 to monitor dynamics
Biochemical characterization:
Differential centrifugation to isolate condensates followed by immunoblotting
Turbidity assays with recombinant BRD3 and RNA
Analysis of concentration-dependent phase separation behaviors
Stimulus-response studies:
Monitor condensate formation under different cellular stresses
Effect of transcriptional inhibitors on BRD3 condensates
Role of post-translational modifications in regulating phase separation
Functional consequences:
ChIP-seq following disruption of phase separation
Transcriptional outputs of genes regulated by BRD3 condensates
Interaction proteomics of proteins recruited to BRD3 condensates
Therapeutic implications:
Screen for compounds that modulate BRD3 phase separation
Evaluate correlation between condensate disruption and gene expression
These approaches can address fundamental questions about how BRD3's phase separation properties contribute to its role in promoting binding to acetylated histones and inducing endoderm gene expression .
Based on recent findings about BRD3's role in regulating inflammatory and stress responses , key methodological approaches include:
Cell-type specific analyses:
Isolation protocols: Standardized methods for obtaining primary cells (e.g., fibroblast-like synoviocytes)
Inflammatory stimulation: Defined conditions using TNF-α, IL-1β, or LPS with time course analysis
Stress induction protocols: Oxidative stress (H₂O₂), heat shock, or nutrient deprivation
BRD3 manipulation strategies:
Genetic approaches: CRISPR/Cas9 knockout, siRNA/shRNA knockdown
Pharmacological approaches: BET inhibitors with varying selectivity profiles
Domain-specific mutants: Bromodomain mutants vs. ET domain mutants
Readout systems:
Cytokine/chemokine production: ELISA, multiplex assays, qRT-PCR
Stress response markers: Western blotting for p62, LC3B (autophagy markers)
Metabolic measurements: Seahorse assays, metabolomics
Temporal considerations:
Acute vs. chronic inflammatory models
Resolution phase analysis
Adaptation mechanisms under prolonged stress
Pathway analysis:
Dissection of signaling cascades (NF-κB, MAPK, etc.)
Intersection with metabolic pathways
Integration with stress response networks
These approaches should incorporate appropriate controls including other BET family members (BRD2, BRD4) to distinguish BRD3-specific effects from general BET protein functions .
Recent advances for studying BRD3-protein interactions include:
Proximity-based labeling techniques:
BioID or TurboID fusion with BRD3 to identify proximal interacting proteins
APEX2-BRD3 fusions for temporal interaction mapping
These methods capture transient interactions often missed by traditional co-IP
Mass spectrometry-based approaches:
Acetylome profiling to identify candidate BRD3-interacting proteins
SILAC or TMT labeling to quantify differential interactions under various conditions
Crosslinking mass spectrometry (XL-MS) to map interaction interfaces
High-resolution microscopy:
Super-resolution imaging to visualize co-localization at sub-diffraction resolution
FRET or FLIM to detect direct protein-protein interactions
Single-molecule tracking to monitor binding dynamics
Biochemical interaction mapping:
Peptide arrays with acetylated and non-acetylated versions of candidate interactors
Surface plasmon resonance or biolayer interferometry for binding kinetics
Hydrogen/deuterium exchange mass spectrometry to map binding interfaces
Functional validation strategies:
Acetylation site mutations in partner proteins
Bromodomain-specific inhibitors to disrupt specific interactions
Domain swapping between BET proteins to identify specificity determinants
These methodologies build upon foundational work showing BRD3's interaction with acetylated GATA1 and GATA2 , enabling more comprehensive characterization of BRD3's non-histone protein interactions.
Seminal studies characterizing BRD3 function include:
GATA1 interaction and hematopoietic regulation:
Lamonica et al. (2011) demonstrated that acetylated GATA1 binds BRD3 in an acetylation-dependent manner to facilitate stable association with chromatin
BRD3 was shown to occupy most GATA1-occupied regulatory DNA elements genome-wide
This study established the role of BRD3 as a "reader" of non-histone acetylated proteins
Chromatin association mechanisms:
Phase separation and gene regulation:
Inflammatory and stress responses:
Cancer connections:
These studies collectively establish BRD3 as a multifunctional chromatin reader with roles in transcriptional regulation, cell differentiation, inflammation, and disease processes.