BRD3 binds acetylated histones (e.g., H3K18ac, H4K12/K16) and transcription factors (e.g., GATA1, RelA) to recruit coactivators and elongation machinery:
Histone binding: BRD3 recognizes acetylated lysines via its bromodomains, promoting chromatin accessibility .
GATA1 interaction: BRD3 binds acetylated GATA1 to regulate erythroid/megakaryocyte gene expression .
Phase separation: BRD3 undergoes liquid-liquid phase separation with lncRNA DIGIT to drive endoderm differentiation .
BRD3 modulates immune pathways in diseases such as rheumatoid arthritis and cancer:
Lymphocyte activation: Upregulated in activated lymphocytes, suggesting a role in adaptive immunity .
Inflammatory gene regulation: Silencing BRD3 in fibroblast-like synoviocytes (FLS) reduces TNF-induced expression of IL-6, MCP-1, and COX2 (Fig. 1) .
Figure 1: BRD3-regulated inflammatory genes in FLS
(Data derived from RNA-seq analysis in TNF-stimulated FLS with BRD3 knockdown)
Gene | Fold Change (BRD3 KD vs Control) |
---|---|
IL-6 | -2.1 |
MCP-1 | -1.8 |
COX2 | -1.6 |
SNAI1 | -1.5 |
BRD3 is implicated in multiple cancers through chromosomal translocations and transcriptional dysregulation:
NUT Midline Carcinoma: BRD3-NUT fusion proteins drive oncogenesis by hijacking BET-dependent transcription .
Colorectal Cancer (CRC): Nuclear BRD3 promotes metastasis via SNAI1/CDC27 upregulation. CRISPR knockout of BRD3, but not BRD4, eradicates CRC metastasis in mice .
Gastric Cancer: Frameshift mutations correlate with reduced PD-L1 expression, altering immune evasion .
BRD3 is detected in synovial macrophages of rheumatoid arthritis (RA) and osteoarthritis (OA) patients:
RA/OA: BRD3 expression correlates with inflammatory scores, suggesting a role in joint inflammation .
Stress Response: BRD3 regulates oxidative stress pathways (e.g., CAT, SOD2) in FLS, though oxidative stress suppresses its expression .
Small-molecule inhibitors (e.g., I-BET151) target conserved bromodomains in BRD2/3/4, displacing them from chromatin:
Cancer Models: BRD3 depletion slows growth in prostate cancer and medulloblastoma, though redundancy with BRD2 limits single-protein targeting .
Immune Regulation: BET inhibitors suppress proinflammatory cytokines (e.g., TNF-α, IL-6) in macrophages and FLS .
Cancer: BRD3-specific inhibitors could overcome resistance to pan-BET inhibitors in CRC and NUT carcinomas .
Autoimmune Diseases: BET inhibition may reduce synovial inflammation in RA by targeting BRD3-expressing macrophages .
BRD3 belongs to the BET family of proteins which includes BRD2, BRD3, BRD4, and BRDT (subfamily V of the bromodomain family). Like other BET proteins, BRD3 contains two highly conserved tandem bromodomains (BD1 and BD2) at the N-terminal end and an extraterminal structural domain at the C-terminal end. The bromodomains exhibit a globular folding pattern with four α-helical tufts (αZ, αA, αB, and αC) and two interhelical loops (ZA and BC) that create a binding pocket for acetylated lysine residues. The conserved asparagine residue in the BC loop forms a hydrogen bond with the acetyl-lysine carbonyl oxygen, which is critical for the recognition and binding of acetylated histones .
BRD3 functions primarily by binding to acetylated lysine residues on chromatin and transcriptional regulators. This binding enables BRD3 to serve as a scaffolding protein in the assembly of multi-protein complexes that regulate diverse biological processes. A key example is BRD3's role in transcription regulation by promoting the binding of transcription factors such as GATA1 to chromatin. Through these interactions, BRD3 influences gene expression patterns and contributes to various cellular functions including cell cycle progression, DNA damage repair, and nuclear reprogramming .
BRD3 is predominantly localized in the nucleus of cells, as confirmed by immunohistochemical analyses of colorectal cancer cells. For effective detection, researchers can employ immunohistochemistry on tissue sections or immunofluorescence in cultured cells using validated anti-BRD3 antibodies. For quantitative analyses, western blotting is useful for measuring protein levels, while chromatin immunoprecipitation (ChIP) can identify genomic regions where BRD3 binds. Single-cell RNA sequencing has also been used to analyze BRD3 expression patterns, revealing that BRD3 is highly expressed in malignant epithelial cells in colorectal cancer tissues .
BRD3 has demonstrated significant involvement in cell cycle regulation, particularly at the G1/S transition. Research using BRD3-knockdown or BRD3-overexpressing colorectal cancer cells has shown that BRD3 suppresses tumor growth by inhibiting the G1/S transition and inducing p21 expression. The p21 protein is a well-known cyclin-dependent kinase inhibitor that blocks cell cycle progression. Single-cell RNA sequencing and spatial transcriptomic analyses have confirmed a positive association between BRD3 expression and high p21 expression in colorectal cancer tissues. Cell cycle checkpoint-related pathways are enriched in epithelial cells with high BRD3 expression, further supporting its role as a cell cycle regulator .
BRD3R, an isoform of BRD3 with reprogramming activity, positively regulates mitosis during nuclear reprogramming. This isoform upregulates a large set of mitotic genes at early stages of reprogramming and associates with mitotic chromatin. Interestingly, many of the mitotic genes upregulated by BRD3R constitute a pluripotent molecular signature, suggesting that mitosis may be a driving force in the reprogramming process. The two BRD3 isoforms demonstrate differential binding to acetylated histones, which may explain their distinct functions in cellular processes. These findings provide a molecular interpretation for the mitotic advantage observed in reprogramming experiments .
BRD3 influences multiple cellular pathways, particularly those related to DNA damage repair responses. Among the top enriched pathways associated with BRD3 and its interactions, DNA damage repair response (DDR) is prominently featured, with 32 out of 42 BRD proteins participating in this process. BRD3 contributes to DDR by helping recognize acetylation signals, recruiting DDR and transcriptional factors, regulating transcription, remodeling chromatin activities, and triggering double-strand break (DSB) repair. Additionally, BRD3 is involved in chromatin remodeling complexes such as the mammalian SWI/SNF complexes, which regulate the accessibility of genomic elements for DNA repair and gene expression .
Recent research has revealed a novel tumor suppressive role for BRD3 in colorectal cancer (CRC). In vitro and in vivo analyses using BRD3-knockdown or BRD3-overexpressing CRC cells demonstrated that BRD3 suppresses tumor growth by inhibiting cell cycle progression at the G1/S transition and inducing p21 expression. Single-cell RNA sequencing of untreated CRC tissues showed that BRD3 is highly expressed in malignant epithelial cells, where cell cycle checkpoint-related pathways are enriched. Spatial transcriptomic and single-cell RNA sequencing analyses confirmed a positive association between BRD3 expression and high p21 expression in CRC tissues. Furthermore, overexpression of BRD3 combined with knockdown of a driver gene in the BRD family showed strong inhibition of CRC cells in vitro .
Clinical analysis of CRC datasets from hospital records and The Cancer Genome Atlas revealed that BET family genes, including BRD3, are overexpressed in colorectal tumor tissues compared to normal tissues. This overexpression pattern suggests complex regulatory mechanisms in cancer, where elevated levels of BRD3 may represent a compensatory response to oncogenic processes. In other cancer types, BRD3 depletion has been shown to slow growth in prostate cancer and medulloblastoma models. Additionally, BRD3 has been implicated in NUT midline carcinoma (NMC), a rare and aggressive cancer characterized by chromosomal rearrangements involving the NUT gene. These findings indicate that BRD3's role may be context-dependent, varying across different cancer types and stages .
Given BRD3's tumor suppressive role in colorectal cancer, BRD3 activation might represent a novel therapeutic approach for this cancer type. Current research has explored combining BRD3 overexpression with knockdown of driver genes in the BRD family, which showed strong inhibition of CRC cells in vitro. While there are several pan-BET inhibitors available, the development of BRD3-specific modulators remains challenging due to the high structural similarity among bromodomains of the BET family, particularly in the acetylated lysine binding pocket. Fragment-based drug discovery approaches have been employed to identify selective BRD3 inhibitors, which may lead to beneficial clinical outcomes with reduced off-target effects compared to pan-BET inhibitors .
Multiple complementary techniques can be employed to study BRD3 protein-protein interactions effectively:
Affinity Purification and Mass Spectrometry: High-throughput approaches for identifying protein complexes associated with BRD3. The BioGRID database (version 4.3.194) contains interaction data derived from various affinity purification methods .
Yeast Two-Hybrid Assays: Useful for identifying direct binary interactions between BRD3 and potential partner proteins .
Co-immunoprecipitation (Co-IP): A widely used method to verify interactions in a cellular context, which can be coupled with western blotting to identify specific interacting partners.
Proximity Ligation Assays: These provide spatial resolution of protein interactions within cells.
Network Analysis Tools: Computational approaches to analyze BRD3 within the broader protein-protein interaction network (PPIN). These analyses can identify dense subgraphs formed by BRD3 and other BRD proteins, revealing topological similarities and functional associations .
To study BRD3's binding to acetylated histones, researchers can employ the following methodologies:
Peptide Binding Assays: Using synthetic acetylated histone peptides to measure binding affinities of purified BRD3 bromodomains.
Surface Plasmon Resonance (SPR): This technique provides quantitative binding kinetics between BRD3 and acetylated histone peptides or proteins.
Isothermal Titration Calorimetry (ITC): Offers thermodynamic parameters of binding interactions between BRD3 and acetylated targets.
Chromatin Immunoprecipitation (ChIP): Identifies genomic regions where BRD3 binds to acetylated histones in vivo.
Structural Studies: X-ray crystallography or NMR spectroscopy can determine the precise molecular interactions between BRD3 bromodomains and acetylated lysine residues. This approach has revealed that a conserved asparagine residue in the BC loop creates a hydrogen bond linking the amide nitrogen and the acetyl-lysine carbonyl oxygen .
Developing selective BRD3 inhibitors presents challenges due to the high structural similarity among BET family bromodomains. Current approaches include:
Fragment-Based Drug Discovery: Screening of fragment libraries against BRD3 to identify novel starting points for inhibitor development. Sygnature Discovery has employed a proprietary fragment library with over 900 compounds to screen against BRD3, focusing on fragments with high novelty and diversity .
Structure-Based Drug Design: Utilizing the crystal structures of BRD3 bromodomains to design selective inhibitors targeting unique structural features.
Differential Binding Analysis: Comparing binding modes across different BET proteins to identify subtle differences that can be exploited for selectivity.
Allosteric Inhibitors: Targeting sites outside the conserved acetyl-lysine binding pocket to achieve selectivity.
Protein Degradation Approaches: PROTAC (PROteolysis TArgeting Chimera) technology that links a BRD3-binding ligand to an E3 ubiquitin ligase recruiting moiety, leading to selective degradation of BRD3 .
Research has identified multiple isoforms of BRD3 with distinct functions. One notable isoform is BRD3R (BRD3 with Reprogramming activity), which plays a critical role in nuclear reprogramming. BRD3R positively regulates mitosis during reprogramming and upregulates a large set of mitotic genes at early stages of the process. The two BRD3 isoforms display differential binding to acetylated histones, which may explain their distinct functions. BRD3R associates with mitotic chromatin and contributes to the mitotic advantage observed in reprogramming. Interestingly, a set of the mitotic genes upregulated by BRD3R constitutes a pluripotent molecular signature, suggesting that BRD3R-mediated mitotic regulation may be a driving force of reprogramming. These findings provide a molecular interpretation for the mitotic advantage observed in traditional reprogramming with somatic cell nuclear transfer (SCNT) .
BRD3 participates in chromatin remodeling complexes that regulate gene expression and DNA repair. One example is the mammalian SWI/SNF (mSWI/SNF) complex, an ATP-dependent chromatin remodeling complex that contains bromodomain modules and regulates the accessibility of genomic elements for DNA damage repair and transcription. While BRD3 itself is not a core component of mSWI/SNF complexes (unlike SMARCA2, SMARCA4, BRD7, and PBRM1), it shares interactome profiles with these proteins, suggesting functional relationships. Network analysis has identified protein complexes where BRD3 interacts with these chromatin remodelers, such as the BRD7-CBP-SWI-SNF complex and the ALL-1 super complex. These interactions allow BRD3 to participate in chromatin remodeling activities, contributing to transcriptional regulation and DNA damage responses .
BRD3, along with other BRD proteins, is integrally involved in DNA damage repair responses (DDR). Among the top enriched pathways associated with BRD proteins, a large set (32 out of 42) are linked to DNA damage repair responses. These proteins participate in:
Recognition of acetylation signals that mark sites of DNA damage
Recruitment of DDR factors and transcriptional regulators to damage sites
Regulation of transcription during the repair process
Chromatin remodeling to facilitate access to damaged DNA
Triggering of double-strand break (DSB) repair mechanisms
BRD3 contributes to these processes through its ability to recognize acetylated lysine residues, which are often present on histones surrounding sites of DNA damage. This recognition enables the recruitment of repair machinery to damaged chromatin. The involvement of BRD3 in DDR highlights its importance in maintaining genomic stability, a critical function that, when dysregulated, can contribute to cancer development .
Protein-protein interaction networks (PPINs) provide valuable insights into BRD3's functional roles. Network analysis of BRD proteins, including BRD3, has established that they serve as hub proteins enriched near the global center of human protein interaction networks, forming an inter-connected PPIN. These analyses have identified dense subgraphs formed by BRD proteins and revealed that different BRD proteins share topological similarity and functional associations. By exploring functional relationships through clustering and Hallmark pathway gene set enrichment analysis, researchers have identified potential biological roles for BRD3 and other BRD proteins. PPIN analysis can predict biological roles for less well-characterized BRD proteins and provide insights into how these proteins function within larger protein complexes. Additionally, PPIN analysis can help identify disease modules and characterize network rewiring events in pathological contexts, potentially pinpointing biologically and therapeutically relevant proteins .
Several contradictions exist in current BRD3 research that require careful consideration:
These contradictions might be resolved through:
Tissue-specific and context-specific studies of BRD3 function
Integration of multiple omics approaches (genomics, proteomics, epigenomics)
Comparison of BRD3 function across different model systems and cancer types
Investigation of post-translational modifications and protein isoforms that might alter BRD3 function
Several emerging technologies hold promise for advancing BRD3 research:
Single-Cell Multi-Omics: Integration of single-cell transcriptomics, proteomics, and epigenomics will provide unprecedented resolution of BRD3 function in heterogeneous cell populations and tissues .
Spatial Transcriptomics and Proteomics: These technologies will allow researchers to map BRD3 expression and activity within the spatial context of tissues, providing insights into its role in specific microenvironments .
CRISPR-Based Epigenome Editing: Precise modification of histone acetylation at specific genomic loci will enable detailed studies of BRD3 recruitment and function.
Cryo-Electron Microscopy: High-resolution structural studies of BRD3 within larger protein complexes will provide insights into its molecular mechanisms.
Protein Degradation Technologies: Targeted degradation approaches like PROTACs will allow selective and rapid depletion of BRD3 to study acute effects on cellular processes.
AI-Driven Drug Discovery: Machine learning approaches will accelerate the development of selective BRD3 modulators by predicting binding affinities and optimizing chemical structures .
Systems Biology Integration: Comprehensive integration of protein interaction, gene expression, and chromatin binding data will provide a holistic view of BRD3 function within cellular networks .
These technologies will help resolve existing contradictions in BRD3 research and reveal new functions and therapeutic opportunities.
Bromodomain-containing proteins (BRD proteins) are a family of proteins that play crucial roles in the regulation of gene expression through their interaction with acetylated lysine residues on histone and non-histone proteins . Among these, Bromodomain Containing 3 (BRD3) is a significant member, known for its involvement in various cellular processes.
BRD3, also known as RING3L, is characterized by the presence of bromodomains, which are evolutionarily conserved protein modules consisting of approximately 110 amino acids . These bromodomains act as specific readers for acetylated lysine residues, facilitating the regulation of transcription, chromatin remodeling, DNA damage repair, and cell proliferation .
BRD3 contains two bromodomains and is part of the bromodomain and extra-terminal (BET) family, which also includes BRD2, BRD4, and BRDT . The BET family proteins are known for their role in recognizing acetylated lysines on histones, thereby influencing gene expression and chromatin structure.
BRD3 functions as a scaffolding protein, assembling multi-protein complexes to regulate diverse biological processes . It is involved in transcriptional regulation by binding to acetylated histones and recruiting other transcriptional machinery components. This interaction is crucial for the activation or repression of specific genes, impacting various cellular functions such as cell cycle control, cell metabolism, and nuclear transport .
Recombinant BRD3 is a form of the protein that is produced through recombinant DNA technology. This involves inserting the gene encoding BRD3 into an expression system, such as E. coli, to produce the protein in large quantities . Recombinant BRD3 is often tagged with a histidine (His) tag to facilitate purification and detection .
BRD3 and other bromodomain-containing proteins have been the focus of extensive research due to their involvement in various diseases, including cancers, inflammation, cardiovascular diseases, and viral infections . Inhibitors targeting BRD3 and other BET proteins have shown promise in preclinical and clinical studies as potential therapeutic agents . These inhibitors work by disrupting the interaction between bromodomains and acetylated lysines, thereby modulating gene expression and offering a novel approach to disease treatment.