BAP1 (BRCA1 Associated Protein 1) is a nuclear protein that functions as a deubiquitinase due to its ubiquitin carboxy-terminal hydrolase (UCH) domain. It is also known by several other names including UCHL2, HUCEP-13, hucep-6, and ubiquitin carboxyl-terminal hydrolase BAP1. Structurally, BAP1 is approximately 80.4 kilodaltons in mass. BAP1 belongs to the polycomb-group proteins (PcGs), which are highly conserved transcriptional repressors required for long-term silencing of genes that regulate various processes including cell fate determination and stem cell pluripotency . As a tumor suppressor, mutations in the BAP1 gene are linked to the development of multiple cancer types, particularly malignant mesothelioma . This critical role in cancer biology makes BAP1 an important target for cancer research, making reliable antibodies essential tools for investigating its expression and function.
BAP1 antibodies are utilized across multiple experimental techniques in molecular and cellular biology research. The most common applications include:
Western Blot (WB): For detecting and quantifying BAP1 protein expression in cell or tissue lysates
Immunohistochemistry (IHC): For visualizing BAP1 expression in formalin-fixed paraffin-embedded (FFPE) tissue sections
Immunoprecipitation (IP): For isolating BAP1 and its binding partners from complex protein mixtures
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of BAP1 in solution
Immunofluorescence (IF): For subcellular localization studies of BAP1 protein
Immunocytochemistry (ICC): For examining BAP1 expression in cultured cells
Flow Cytometry (FCM): For analyzing BAP1 expression at the single-cell level
The selection of application depends on your specific research question, with different antibodies being optimized for different applications as indicated in supplier information.
Selecting the optimal BAP1 antibody requires consideration of several key factors:
Target epitope: Determine whether you need an antibody targeting a specific region of BAP1 (N-terminal, C-terminal, or internal domain). This is particularly important if you're studying specific isoforms or if post-translational modifications may affect antibody binding .
Validated applications: Review the applications for which the antibody has been validated. For example, some antibodies may perform well in Western blot but poorly in IHC, or vice versa .
Species reactivity: Ensure the antibody recognizes BAP1 in your species of interest. While many antibodies target human BAP1, cross-reactivity with mouse, rat, or other species varies significantly between products .
Clonality: Polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variability. Monoclonal antibodies provide high specificity and consistency but might be less robust to changes in protein conformation .
Published validation: Review citations and figures provided by manufacturers showing actual experimental results with the antibody .
Conjugation needs: Determine if you require an unconjugated antibody or one with a specific tag (biotin, FITC, HRP, etc.) based on your detection method .
Always pilot test multiple antibodies if possible, especially for critical experiments, to identify which performs best in your specific experimental system.
Thorough validation of a BAP1 antibody is crucial before using it for definitive experiments. Follow these methodological steps for comprehensive validation:
Positive and negative controls: Include known BAP1-positive tissue/cells and compare with BAP1-knockout or BAP1-depleted samples. For human samples, mesothelioma cell lines with known BAP1 status serve as excellent controls .
Western blot validation: Verify the antibody detects a band of the expected molecular weight (~80.4 kDa). Check for non-specific bands that could interfere with interpretation .
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide to confirm specificity of binding. The specific signal should be significantly reduced or eliminated.
Multiple techniques confirmation: If planning to use the antibody for IHC, confirm the specificity by also performing Western blot or immunofluorescence on the same samples.
siRNA/shRNA knockdown: Demonstrate reduced signal intensity following BAP1 knockdown, confirming the antibody is truly detecting BAP1.
Titration experiments: Perform dilution series to identify the optimal antibody concentration that maximizes specific signal while minimizing background.
Cross-reactivity assessment: If working across species, validate the antibody in each species separately, as cross-reactivity efficiency often varies despite manufacturer claims .
Reproducibility testing: Perform replicate experiments to ensure consistent results across different days and sample preparations.
Document all validation steps meticulously, as this information will strengthen the reliability of your research findings.
Optimizing BAP1 immunohistochemistry protocols for different tissue types requires systematic adjustment of several parameters:
Antigen retrieval method: BAP1 detection often requires heat-induced epitope retrieval (HIER). Test both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) to determine which provides optimal staining for your specific tissue. Some BAP1 epitopes may be particularly sensitive to pH during retrieval .
Fixation considerations: Overfixation can mask BAP1 epitopes. For FFPE tissues, fixation in 10% neutral buffered formalin for 24-48 hours is generally recommended. For challenging samples, extend the antigen retrieval time.
Antibody concentration and incubation conditions: Titrate the primary antibody concentration specifically for each tissue type. Nuclear proteins like BAP1 may require longer incubation times (overnight at 4°C) for optimal penetration and binding .
Detection system sensitivity: For tissues with low BAP1 expression, use amplification systems such as polymer-based detection methods or tyramide signal amplification.
Tissue-specific blocking: Certain tissues (particularly liver, kidney, and lymphoid tissues) may require additional blocking steps to reduce background. Test 5-10% normal serum from the species of the secondary antibody plus 1% BSA.
Counterstaining adjustment: Optimize hematoxylin counterstaining time to ensure nuclear details remain visible without obscuring the BAP1 nuclear signal.
Automated vs. manual staining: Compare results between automated staining platforms and manual methods, as some antibodies perform differently in each system.
Tissue-specific positive controls: Include known positive controls of the same tissue type you're investigating, as BAP1 expression patterns may vary between tissues.
Document all optimization parameters in your protocol to ensure reproducibility across experiments and between laboratory members.
Several challenges can arise when using BAP1 antibodies for Western blotting, and awareness of these potential pitfalls can help ensure reliable results:
Protein extraction efficiency: BAP1 is a nuclear protein, requiring efficient nuclear extraction protocols. Inadequate extraction can lead to falsely decreased BAP1 signal. Use nuclear extraction buffers containing DNase to break down chromatin and release nuclear proteins effectively .
Sample preparation artifacts: Repeated freeze-thaw cycles can degrade BAP1. Prepare fresh lysates or add additional protease inhibitors if using stored samples.
Transfer efficiency issues: BAP1's relatively large size (~80.4 kDa) may require optimization of transfer conditions. Use lower percentage polyacrylamide gels (8-10%) and adjust transfer time/voltage accordingly .
Non-specific banding patterns: Some BAP1 antibodies may recognize splice variants or cross-react with related deubiquitinases. Always include positive control lysates with known BAP1 expression to identify the correct band .
Antibody concentration optimization: Under-diluted antibodies can lead to high background, while over-dilution results in weak specific signal. Titrate antibody concentration for each new lot.
Detection system sensitivity: For samples with low BAP1 expression, enhanced chemiluminescence (ECL) detection may not be sensitive enough. Consider using more sensitive detection reagents such as ECL Plus or SuperSignal West Femto.
Blocking optimization: Milk-based blocking solutions may contain phosphatases that interfere with phospho-specific BAP1 detection. Use BSA-based blocking solutions when analyzing phosphorylated forms of BAP1.
Loading control selection: Standard housekeeping proteins may not accurately reflect nuclear protein loading. Consider using nuclear-specific loading controls such as Lamin B1 or HDAC1.
By systematically addressing these potential issues, you can develop a robust Western blotting protocol for reliable BAP1 detection.
Designing experiments to investigate BAP1's protein-protein interactions requires a multi-technique approach:
Co-immunoprecipitation (Co-IP): Use BAP1 antibodies suitable for immunoprecipitation to pull down BAP1 and its binding partners. Consider both forward and reverse Co-IP (immunoprecipitate with anti-BAP1 and with antibodies against suspected interacting partners) .
Proximity ligation assay (PLA): For detecting protein interactions in situ, PLA allows visualization of protein complexes within cells with high sensitivity and specificity. This is particularly useful for confirming interactions in their native cellular context.
Bimolecular fluorescence complementation (BiFC): By tagging BAP1 and potential interacting proteins with complementary fragments of a fluorescent protein, interaction can be visualized when the fragments come together to form a functional fluorophore.
Cell fractionation analysis: Since BAP1 is primarily nuclear, separate nuclear and cytoplasmic fractions to determine where interactions occur. Some interactions may be compartment-specific or may change BAP1's localization.
Sequential immunoprecipitation: For complex formation studies, perform tandem immunoprecipitations to isolate specific multi-protein complexes containing BAP1.
Crosslinking approaches: Prior to immunoprecipitation, use protein crosslinkers to stabilize transient interactions that might be lost during standard Co-IP procedures.
Truncation and mutation analysis: Create BAP1 constructs with specific domains deleted or mutated to map the regions required for particular protein interactions.
Competitive peptide disruption: Use peptides corresponding to potential binding interfaces to competitively disrupt specific interactions, confirming their importance.
These approaches can be combined to build a comprehensive understanding of BAP1's interaction network and how these interactions influence its tumor suppressor functions.
Studying post-translational modifications (PTMs) of BAP1 requires specific methodological approaches:
Phospho-specific antibodies: Several suppliers offer antibodies targeting specific phosphorylation sites of BAP1, such as Ser592 . These allow direct detection of phosphorylated BAP1 in Western blotting, immunohistochemistry, or immunofluorescence.
Phosphatase treatment controls: To confirm phospho-specific antibody specificity, treat duplicate samples with lambda phosphatase and observe loss of signal with phospho-specific antibodies while total BAP1 detection remains unchanged.
Phos-tag SDS-PAGE: This technique incorporates Phos-tag molecules into polyacrylamide gels, causing phosphorylated proteins to migrate more slowly. This allows separation of phosphorylated and non-phosphorylated BAP1 without phospho-specific antibodies.
Mass spectrometry approach: For comprehensive PTM mapping, immunoprecipitate BAP1 and analyze by mass spectrometry. This can identify multiple modification sites simultaneously, including phosphorylation, ubiquitination, SUMOylation, and acetylation.
PTM site mutagenesis: Create BAP1 constructs with mutations at potential modification sites (changing serines/threonines to alanines for phosphorylation studies, or lysines to arginines for ubiquitination studies) to assess functional consequences.
PTM induction experiments: Treat cells with PTM-inducing conditions (e.g., cell cycle synchronization, DNA damage, hypoxia) to study dynamic regulation of BAP1 modifications.
Sequential immunoprecipitation: For studying multi-modified BAP1, perform sequential IPs first with PTM-specific antibodies followed by BAP1 antibodies, or vice versa.
Proximity ligation assays: Use antibodies against BAP1 and specific PTMs to visualize modified BAP1 in situ with high specificity and sensitivity.
These techniques can provide insights into how PTMs regulate BAP1's tumor suppressor function, subcellular localization, and protein-protein interactions.
Differentiating between wild-type and mutant BAP1 in cancer samples requires strategic experimental approaches:
Immunohistochemistry pattern analysis: Wild-type BAP1 typically shows strong nuclear localization. Loss of nuclear staining with retained cytoplasmic staining, or complete absence of staining, often indicates inactivating BAP1 mutations. Develop a scoring system considering both intensity and subcellular localization .
Mutation-specific antibodies: For recurrent mutations, consider developing or sourcing antibodies that specifically recognize common BAP1 mutants. These are particularly valuable for missense mutations that may retain protein expression but lose function.
Western blot mobility analysis: Some mutations (particularly insertions/deletions) may alter the apparent molecular weight of BAP1. High-resolution Western blotting can detect these mobility shifts.
Combined genomic-proteomic approach: Correlate antibody staining patterns with genomic sequencing data from the same samples to establish reliable IHC criteria for predicting mutation status.
C-terminal vs. N-terminal antibodies: Since many truncating mutations affect the C-terminus, compare staining patterns using antibodies targeting different regions of BAP1 . Discrepancies between N- and C-terminal antibody staining may indicate truncating mutations.
Functional readouts: Use antibodies against BAP1 substrates or downstream targets as surrogate markers for BAP1 activity, which may distinguish functional from non-functional BAP1 even when the protein is expressed.
Quantitative image analysis: Employ digital image analysis of IHC slides to quantify nuclear-to-cytoplasmic ratios of BAP1 staining, potentially identifying subtle localization changes in certain mutants.
Tissue microarray validation: Validate your approach using tissue microarrays containing samples with known BAP1 mutation status to establish sensitivity and specificity of your antibody-based detection method.
This multi-faceted approach can provide valuable diagnostic and prognostic information for cancer samples, particularly for mesotheliomas where BAP1 status has important clinical implications .
BAP1 antibodies serve as critical tools for evaluating its tumor suppressor activity across various cancer types:
Comparative expression analysis: Quantify BAP1 protein levels in matched tumor and normal adjacent tissues using immunohistochemistry or Western blotting. Progressive loss of BAP1 expression often correlates with increasing tumor grade or stage .
Prognostic biomarker development: Perform large-scale tissue microarray studies with clinical outcome data to correlate BAP1 expression patterns with patient survival, treatment response, and recurrence rates. This requires standardized staining and scoring protocols using validated BAP1 antibodies .
Mechanistic pathway studies: Combine BAP1 antibodies with antibodies against known downstream targets to map pathway disruptions in specific cancer contexts. This might include PRC1/2 complex components or DNA damage response proteins.
Synthetic lethality screening: Use BAP1 antibodies to confirm BAP1 status in cell lines before performing drug or genetic screens to identify vulnerabilities created by BAP1 loss. Western blot validation is essential to confirm complete protein loss.
Tumor heterogeneity mapping: Apply multiplex immunofluorescence with BAP1 and other marker antibodies to analyze intratumoral heterogeneity of BAP1 expression and identify subpopulations with differential BAP1 status.
Circulating tumor cell analysis: Develop protocols using BAP1 antibodies to assess BAP1 status in circulating tumor cells, potentially providing a liquid biopsy approach for monitoring disease.
Therapeutic response prediction: Correlate pre-treatment BAP1 expression (by IHC or Western blot) with response to specific therapies to develop predictive biomarkers for personalized treatment approaches.
Cancer evolution studies: Use BAP1 antibodies to track changes in BAP1 expression from pre-malignant lesions through primary tumors to metastases, providing insights into the timing and impact of BAP1 loss during tumor progression.
These approaches collectively enhance our understanding of BAP1's role in tumorigenesis and can inform the development of targeted therapeutic strategies.
Investigating BAP1's functions in DNA damage response requires specialized experimental approaches:
Immunofluorescence co-localization: Use BAP1 antibodies together with antibodies against DNA damage markers (γH2AX, 53BP1, RAD51) to assess BAP1 recruitment to DNA damage sites after induction with UV, ionizing radiation, or chemical agents. Time-course experiments can reveal the dynamics of this recruitment .
Chromatin immunoprecipitation (ChIP): Employ BAP1 antibodies suitable for ChIP to determine whether BAP1 is recruited to specific genomic loci following DNA damage. This approach can be combined with sequencing (ChIP-seq) for genome-wide analysis.
Proximity-based labeling: Use techniques like BioID or APEX2 fused to BAP1 to identify proteins that interact with BAP1 specifically after DNA damage, providing insights into its damage-specific interactome.
FRAP analysis: Perform fluorescence recovery after photobleaching with GFP-tagged BAP1 and validate findings with antibody staining of endogenous BAP1 to assess the dynamics of BAP1 mobility after DNA damage.
Cell cycle-resolved analysis: Combine BAP1 immunostaining with cell cycle markers or synchronization protocols to determine whether BAP1's role in DNA damage response varies throughout the cell cycle.
Ubiquitination dynamics: Use antibodies against ubiquitin along with BAP1 to investigate whether BAP1's deubiquitinase activity modifies specific substrates following DNA damage.
Laser microirradiation: Combine this technique with live-cell imaging and subsequent fixation and immunostaining with BAP1 antibodies to visualize recruitment to damage sites with high spatial and temporal resolution.
Functional recovery assays: Measure DNA repair efficiency (comet assay, repair reporter assays) in cells with different BAP1 status, confirmed by antibody detection, to quantify BAP1's contribution to specific repair pathways.
These approaches collectively provide a comprehensive understanding of how BAP1 participates in maintaining genomic integrity through DNA damage response mechanisms.
Designing experiments to elucidate BAP1's functions in cell cycle regulation requires multiple complementary approaches:
Synchronized cell population analysis: Synchronize cells at different cell cycle phases using methods like double thymidine block or nocodazole treatment. Use BAP1 antibodies to assess its expression, localization, and post-translational modifications across cell cycle stages by Western blotting and immunofluorescence .
Flow cytometry with BAP1 co-staining: Combine DNA content analysis (propidium iodide staining) with BAP1 immunostaining for flow cytometry to correlate BAP1 levels with cell cycle position at the single-cell level.
Cell cycle marker co-localization: Perform double immunofluorescence with BAP1 antibodies and antibodies against phase-specific markers (cyclin D1, cyclin E, cyclin B1, phospho-histone H3) to assess potential co-localization or mutual exclusivity patterns.
CDK-dependent phosphorylation analysis: Use phospho-specific BAP1 antibodies to determine whether BAP1 is phosphorylated by cell cycle-dependent kinases at specific phases .
Chromatin association dynamics: Perform biochemical fractionation followed by Western blotting with BAP1 antibodies to assess whether BAP1's association with chromatin changes during cell cycle progression.
Cell cycle protein stability assays: Use cycloheximide chase experiments with BAP1 antibody detection to determine if BAP1 stability varies across the cell cycle.
BAP1 substrate identification: Combine BAP1 manipulation (overexpression, depletion, or mutation) with antibody-based detection of cell cycle regulators to identify potential substrates whose ubiquitination status changes in a cell cycle-dependent manner.
Live cell imaging validation: After establishing key findings with fixed-cell antibody-based methods, validate with live-cell approaches using fluorescently tagged BAP1, with antibody staining as confirmation.
These methodologies provide complementary data on how BAP1 may regulate or be regulated by the cell cycle machinery, offering insights into another facet of its tumor suppressor function.
BAP1 antibodies can be integrated into cutting-edge single-cell analysis techniques with these methodological approaches:
Single-cell Western blotting: Adapt traditional Western blot techniques for single-cell resolution using specialized platforms (e.g., Milo, ProteinSimple). Validate BAP1 antibodies specifically for these platforms, as sensitivity requirements differ from traditional Western blotting .
Mass cytometry (CyTOF): Conjugate BAP1 antibodies with heavy metal isotopes for inclusion in CyTOF panels. This allows simultaneous detection of BAP1 alongside dozens of other proteins at single-cell resolution without fluorescence spillover concerns.
scRNA-seq with protein detection: Integrate BAP1 antibodies into CITE-seq or REAP-seq protocols, which allow simultaneous measurement of transcriptome and selected proteins in single cells. This reveals correlations between BAP1 protein levels and gene expression programs.
Microfluidic antibody capture: Use BAP1 antibodies in microfluidic devices designed for single-cell protein secretion assays, adapting protocols to detect intracellular BAP1 after cell permeabilization.
Imaging mass cytometry: Apply metal-conjugated BAP1 antibodies for highly multiplexed tissue imaging at subcellular resolution, allowing spatial analysis of BAP1 in relation to multiple cell type markers and functional proteins.
Digital spatial profiling: Incorporate BAP1 antibodies into spatial profiling platforms that capture proteins from specific tissue regions, enabling spatial analysis of BAP1 expression patterns within the tumor microenvironment.
Single-cell ChIP-seq: Adapt BAP1 antibodies for use in single-cell chromatin immunoprecipitation protocols to assess cell-to-cell variation in BAP1 chromatin binding patterns.
Phospho-flow cytometry: For studying BAP1 phosphorylation in single cells, optimize phospho-specific BAP1 antibodies for flow cytometry applications with appropriate fixation and permeabilization protocols .
These advanced applications enable researchers to move beyond population averages to understand cell-to-cell heterogeneity in BAP1 expression, localization, and function.
When investigating BAP1 in non-human model organisms, several important considerations must be addressed:
Sequence homology verification: Before selecting an antibody, perform sequence alignment between human BAP1 and the model organism's ortholog. Many commercially available antibodies are raised against human BAP1 and may have limited cross-reactivity despite manufacturer claims .
Species-specific validation: Thoroughly validate any antibody claimed to be cross-reactive with your model organism. Include positive controls (tissue known to express BAP1) and negative controls (BAP1 knockout or knockdown samples) from your specific model organism .
Epitope conservation analysis: Identify the specific epitope recognized by the antibody and assess its conservation in your model organism. Antibodies targeting highly conserved regions generally offer better cross-reactivity .
Application-specific testing: An antibody that cross-reacts in Western blot may fail in immunohistochemistry or immunoprecipitation applications. Validate for each specific application in your model organism .
Protocol optimization: Fixation, antigen retrieval, and blocking conditions often need significant modification when moving between species. Systematic optimization is essential for reliable results.
Paralog consideration: Some model organisms have multiple BAP1 paralogs or related deubiquitinases. Ensure your antibody specifically recognizes the intended target without cross-reactivity to related proteins.
Expression pattern verification: Use complementary methods such as in situ hybridization or reporter constructs to confirm that antibody staining patterns match expected BAP1 expression domains in your model organism.
Functional conservation assessment: Before extensive antibody-based studies, confirm that BAP1 functions are conserved in your model organism through genetic or biochemical approaches, as this affects interpretation of your findings.
Careful attention to these considerations will ensure reliable detection of BAP1 in model organisms, allowing valid cross-species comparisons and translational insights.
Designing effective multiplex immunoassays that incorporate BAP1 detection requires careful consideration of several technical factors:
Antibody compatibility assessment: Test BAP1 antibodies with other antibodies in your planned panel to identify potential cross-reactivity issues. Perform single-staining controls before attempting multiplexing .
Epitope retrieval optimization: When multiple proteins require different antigen retrieval conditions, conduct systematic testing to find a compromise condition that adequately retrieves BAP1 epitopes without compromising detection of other targets .
Signal amplification strategies: For targets with disparate expression levels, implement differential signal amplification. For example, use tyramide signal amplification specifically for low-abundance targets while using standard detection for highly expressed proteins including BAP1.
Fluorophore selection for spectral separation: When designing multiplex immunofluorescence panels including BAP1, select fluorophores with minimal spectral overlap and use appropriate controls for spectral unmixing.
Sequential staining protocols: For challenging combinations, develop sequential staining protocols with heat or chemical inactivation steps between rounds. Validate that BAP1 antibody staining is not affected by these intermediate treatments.
Antibody conjugation considerations: If directly conjugating BAP1 antibodies, test multiple conjugation chemistries and fluorophore-to-antibody ratios to maintain affinity and specificity while maximizing signal strength.
Multiplex validation strategies: Validate multiplex results against single-plex controls to ensure that sensitivity and specificity for BAP1 detection remain consistent in the multiplex context.
Data normalization approaches: Develop robust normalization strategies for quantitative multiplex assays, particularly important when comparing BAP1 levels across different specimens or experimental conditions.
These methodological considerations will help develop reliable multiplex assays incorporating BAP1 detection, enabling complex analyses of pathway interactions and cellular heterogeneity in normal and disease contexts.
The field of BAP1 antibody-based research continues to evolve, with several notable emerging trends:
Integration with spatial biology platforms: BAP1 antibodies are increasingly being incorporated into highly multiplexed spatial profiling technologies such as imaging mass cytometry, Codex, and digital spatial profiling. These approaches provide unprecedented insights into BAP1's role within the spatial context of tissues and tumor microenvironments .
Single-cell resolution techniques: Moving beyond bulk tissue analysis, researchers are adapting BAP1 antibodies for single-cell Western blotting, mass cytometry, and combined protein-transcriptome analysis at single-cell resolution, revealing previously unappreciated heterogeneity in BAP1 expression and function .
Enhanced validation standards: The field is moving toward more rigorous validation of BAP1 antibodies, including the use of CRISPR-engineered knockout controls and assessment across multiple applications and fixation conditions. This trend is improving data reproducibility and reliability .
PTM-specific detection: Development and application of antibodies targeting specific post-translational modifications of BAP1 are providing new insights into its regulation. Phospho-BAP1 antibodies, in particular, are enabling studies of how BAP1 is dynamically regulated during cell cycle progression and in response to cellular stresses .
Automation and standardization: To improve reproducibility in diagnostic applications, automated staining platforms with validated BAP1 antibody protocols are being developed and implemented in clinical research settings .
Combination with functional genomics: Researchers are combining antibody-based detection of BAP1 with CRISPR screens, creating powerful platforms for identifying synthetic lethal interactions and potential therapeutic vulnerabilities in BAP1-deficient cancers.
Clinical biomarker development: BAP1 antibodies are being rigorously evaluated as diagnostic, prognostic, and predictive biomarkers across multiple cancer types, with standardized immunohistochemical protocols moving toward clinical implementation .
Integrative multi-omics approaches: BAP1 antibody-based proteomics is being integrated with genomics, transcriptomics, and epigenomics to build comprehensive models of BAP1 function in health and disease.