RBBP8 antibodies are immunological reagents designed to detect and quantify the RBBP8 protein in experimental settings. These antibodies enable researchers to investigate RBBP8's roles in homologous recombination (HR) repair, cell cycle progression (S/G2 phase), and interactions with partners like BRCA1, ATM, and RAD51 .
RBBP8 facilitates DNA double-strand break (DSB) repair by promoting resection of DNA ends, enabling HR repair in S/G2-phase cells .
Knockdown of RBBP8 in gastric cancer cells (AGS, N87) increases DNA damage markers (γ-H2AX) and sensitizes cells to PARP inhibitors, suggesting synthetic lethality .
RBBP8 modulates the G1/S transition by suppressing P21 transcription via chromatin remodeling, enabling progression into phases permissive for HR repair .
CRISPR screens revealed that RBBP8 deletion arrests cells in S/G2 phase and reduces protein synthesis under endoplasmic reticulum (ER) stress .
Cancer Therapy: RBBP8 overexpression correlates with chemotherapy resistance in gastric cancer, making it a potential biomarker for PARP inhibitor efficacy .
Liver Disease: RBBP8 deficiency protects hepatocytes from ER stress-induced damage by attenuating ATF4 activation .
Western Blot Protocol: Optimized dilution (1:500–1:1000) with detection in nuclear extracts .
Functional Assays: siRNA-mediated knockdown in HEK293T cells confirmed reduced ATF4 and XBP1s levels under ER stress .
Biomarker Potential: High RBBP8 expression in liver cancer correlates with ATF4 activation, indicating a role in stress adaptation .
PARP Inhibitor Synergy: Combining RBBP8 inhibition with PARP blockers enhances cytotoxicity in gastric cancer models .
Ongoing research aims to elucidate:
The structural basis of RBBP8’s interaction with BRCA1 and the MRN complex.
Tissue-specific roles of RBBP8 in tumorigenesis and chemoresistance.
RBBP8 (also known as CTIP, CtBP-interacting protein, retinoblastoma-binding protein 8) is a DNA endonuclease that plays crucial roles in homologous recombination (HR) repair of DNA damage. The protein is particularly important in DNA terminal excision during double-strand break repair processes . RBBP8's significance extends to its involvement in cell cycle regulation, as its phosphorylation increases when cells cross the G1/S phase and maintains high levels during S/G2 phase . Recent research has also revealed RBBP8's role in activating ATF4, linking DNA damage stress with unfolded protein response (UPR) activation . These multiple functions make RBBP8 antibodies valuable tools for studying DNA damage repair mechanisms, cell cycle regulation, and stress response pathways.
When selecting an RBBP8 antibody, researchers should consider:
Species reactivity: Confirm the antibody reacts with your species of interest (e.g., human, mouse)
Host species: Consider the host (e.g., rabbit) for compatibility with your secondary detection systems and to avoid cross-reactivity in multi-labeling experiments
Validated applications: Ensure the antibody is validated for your specific application (ELISA, IF, IHC, ICC, WB)
Epitope location: For phosphorylation studies, select antibodies that target specific phosphorylation sites relevant to cell cycle phases
Validation evidence: Review immunohistochemistry and western blot validation images provided by manufacturers to confirm specificity
Clonality: Monoclonal antibodies offer higher specificity while polyclonal antibodies may provide stronger signals
For optimal Western blot results with RBBP8 antibodies:
Sample preparation: Prepare protein samples at a ratio of 40 μg: 20 μL
Electrophoresis conditions: Run concentrated gel at 80V for 30 min, followed by separated gel at 100V for 60 min
Transfer parameters: Perform transfer at 390V for 70 min for efficient transfer of high molecular weight RBBP8 (101942 MW)
Blocking: Use 5% skimmed milk for 1-2 hours at room temperature
Antibody incubation: Incubate primary antibody overnight at 4°C, followed by secondary antibody at room temperature
Controls: Include negative controls and, if possible, RBBP8 knockdown samples to confirm specificity
Visualization: For cell cycle-dependent phosphorylation studies, consider using phos-tag gels to better separate phosphorylated forms
For immunofluorescence studies examining RBBP8's role in DNA damage:
Experimental groups to consider: Include negative control, PARP inhibitor treatment, RBBP8 knockdown, RBBP8 knockdown + PARP inhibitor, and RBBP8 overexpression groups
Co-staining markers: Co-stain with γ-H2AX (DNA damage marker) using Alexa Fluor 568 goat anti-rabbit secondary antibody
RAD51 activation visualization: Use Alexa Fluor 488 goat anti-mouse for detecting RAD51 foci formation
BrdU incorporation: For single-stranded DNA formation studies, incubate cells with BrdU and visualize using Alexa Fluor 488 goat anti-mouse
Microscopy: Use confocal microscopy for optimal detection of nuclear foci
Quantification: Count the number of RAD51 or γ-H2AX foci per nucleus across multiple fields (minimum 100 cells) to ensure statistical significance
RBBP8 phosphorylation and function are highly cell cycle-dependent, requiring proper synchronization methods:
G1 phase synchronization: Implement TdR double blocking protocol
Verification: Confirm cell cycle phase distribution using flow cytometry with propidium iodide staining
RBBP8 phosphorylation detection: Use phospho-specific antibodies or general RBBP8 antibodies combined with phosphatase treatments to confirm phosphorylation status
Timing considerations: For optimal detection of phosphorylated RBBP8, collect samples during S/G2 phase when phosphorylation levels are highest
Controls: Include asynchronous cells as controls and verify synchronization efficiency before proceeding with RBBP8 analysis
Discrepancies in RBBP8 detection may arise from:
Phosphorylation states: RBBP8 undergoes cell cycle-dependent phosphorylation, which can affect antibody binding and protein mobility in gels
Protein interactions: RBBP8 forms complexes with other proteins which may mask epitopes in certain contexts
Alternative splicing: Different isoforms may be detected differentially by antibodies targeting different regions
Subcellular localization: RBBP8 primarily localizes to the nucleus, but improper sample preparation may affect detection
Fixation sensitivity: For immunohistochemistry and immunofluorescence, different fixation methods may alter epitope accessibility
To address these issues:
Use multiple antibodies targeting different epitopes
Include appropriate controls (tissue-specific positive controls, knockdown negative controls)
Combine multiple detection methods (IF, WB, IP) to confirm results
Consider cell cycle phase when interpreting results
When encountering inconsistent RBBP8 staining in tissue samples:
Antigen retrieval optimization: Test multiple antigen retrieval methods as RBBP8 epitopes may be sensitive to specific retrieval conditions
Blocking protocol adjustment: Increase blocking time or try alternative blocking agents to reduce non-specific binding
Antibody titration: Perform careful antibody titration to determine optimal concentration for specific tissues
Positive controls: Include known positive control tissues (e.g., human lung carcinoma tissue has been validated)
Peptide blocking: Perform peptide blocking experiments to confirm specificity, as demonstrated in validation images
Sample processing standardization: Ensure consistent fixation times and processing methods across samples
Multi-antibody validation: Use multiple antibodies against different RBBP8 epitopes to confirm staining patterns
For investigating synthetic lethality involving RBBP8:
Experimental design:
Detection methods:
Data analysis:
This approach has successfully demonstrated synthetic lethality between RBBP8 knockdown and PARP inhibition in gastric cancer cell lines, with significant inhibition of cell viability and promotion of apoptosis .
Recent CRISPR screen studies have identified RBBP8's novel role in UPR activation . To investigate this connection:
Reporter systems:
Develop or utilize ATF4 reporter-based cell models to monitor UPR activation
Design dual reporter systems to simultaneously track DNA damage and UPR activation
Stress induction protocols:
Readout measurements:
Validation approaches:
This growing research area suggests a previously unrecognized role for RBBP8 in linking nuclear DNA damage stress with endoplasmic reticulum stress responses .
RBBP8 phosphorylation is critical to its function and highly cell cycle-dependent . Advanced approaches include:
Phospho-specific antibodies:
Use antibodies targeting specific RBBP8 phosphorylation sites
Validate specificity using phosphatase treatment controls
Compare signals between different cell cycle phases
Phos-tag SDS-PAGE:
Incorporate Phos-tag into polyacrylamide gels to enhance separation of phosphorylated proteins
Detect mobility shifts corresponding to different phosphorylation states
Quantify the ratio of phosphorylated to non-phosphorylated forms
Mass spectrometry approaches:
Employ phospho-enrichment protocols prior to MS analysis
Map specific phosphorylation sites across the RBBP8 protein
Perform quantitative analysis to determine relative phosphorylation levels
Cell cycle synchronization:
Implement precise synchronization at G1/S boundary and throughout S/G2 phases
Collect time-course samples to track phosphorylation dynamics
Correlate phosphorylation patterns with functional outcomes in DNA repair
| Cell Cycle Phase | RBBP8 Phosphorylation Status | Functional Implications | Detection Method |
|---|---|---|---|
| G1 | Minimal phosphorylation | Limited HR repair activity | Western blot, IF |
| S | Increasing phosphorylation | Active DNA end resection | Phos-tag SDS-PAGE, MS |
| G2 | Maximal phosphorylation | Peak HR repair capacity | Phospho-specific antibodies |
| M | Decreasing phosphorylation | Reduced repair activity | Western blot with mobility shift |
These approaches enable researchers to correlate RBBP8 phosphorylation states with its functional roles in homologous recombination repair throughout the cell cycle .
While RBBP8 antibodies are intended for research use only , exploratory translational studies may consider:
Tissue microarray (TMA) analysis:
Standardize staining protocols for consistent results across patient samples
Use digital pathology and automated scoring to quantify expression levels
Correlate with clinical parameters and outcomes
Expression correlation approaches:
Functional assessment:
Evaluate DNA damage repair capacity in patient-derived samples
Assess correlation between RBBP8 expression and response to DNA-damaging therapies
Consider potential as a biomarker for PARP inhibitor sensitivity
Bioinformatic validation:
Analyze public datasets to validate expression patterns across cancer types
Perform survival analysis stratified by RBBP8 expression levels
Integrate multi-omics data to understand regulatory mechanisms
Note: Any potential diagnostic applications would require extensive validation and regulatory approval beyond the current research-use status of available antibodies .
For researchers exploring RBBP8 as a therapeutic target:
Target validation approaches:
Confirm synthetic lethality effect through multiple cell line models
Validate in 3D organoid cultures and xenograft models
Assess potential toxicity in normal cells and tissues
Combination therapy design:
Test RBBP8 intervention in combination with PARP inhibitors at various concentrations
Determine optimal dosing schedules (concurrent vs. sequential)
Evaluate potential for resistance development through long-term studies
Molecular intervention strategies:
Compare efficacy of different RBBP8 targeting approaches (siRNA, small molecule inhibitors)
Design selective inhibitors that target specific functional domains
Consider developing inhibitors that specifically block RBBP8's interaction with DNA or partner proteins
Biomarker development:
Identify predictive biomarkers for response to RBBP8-targeted therapy
Develop companion diagnostic approaches
Consider genomic and proteomic signatures beyond RBBP8 expression alone