BCL7C is a component of the SWI/SNF chromatin remodeling complex (GBAF subcomplex), which regulates DNA-histone interactions in an ATP-dependent manner . Functional studies reveal:
Anti-apoptotic Activity: Early reports suggested a role in inhibiting apoptosis .
Tumor-Suppressive Role: Contradicting earlier claims, recent studies demonstrate BCL7C suppresses ovarian cancer by binding mutant p53 (mtp53), inhibiting its oncogenic activity .
BCL7C’s tumor-suppressive role is best characterized in ovarian cancer:
Interaction with Mutant p53: BCL7C binds mtp53 (e.g., R175H, R273H), blocking its transcriptional activation of pro-tumorigenic genes .
Impact on Cell Behavior:
Clinical Correlation: Low BCL7C levels correlate with poor prognosis in ovarian carcinomas and other cancers .
Recombinant BCL7C (e.g., PRO-1781, ab174409) is produced in E. coli and used for:
Disease Associations:
Prognostic Marker: Underexpression predicts unfavorable outcomes in ovarian cancer .
BCL7C (B-cell CLL/lymphoma 7 protein family member C) is located on chromosome 16p11.2 and shares partial sequence homology with two other family members, BCL7A and BCL7B. While research on BCL7C is still emerging, it has been identified as having significant roles in cancer biology, particularly as a tumor suppressor in ovarian cancer .
Methodological approach: Genomic characterization of BCL7C typically involves sequencing analysis, expression profiling across tissue types, and comparative genomic alignment with other BCL7 family members to identify conserved domains and unique regions.
Current research data on BCL7C tissue-specific expression patterns in normal tissues is limited. Understanding BCL7C expression in non-pathological states provides crucial context for interpreting its altered expression in disease.
Methodological approach: Researchers should employ a combination of RNA-seq, immunohistochemistry with validated antibodies, and tissue microarrays to establish comprehensive expression profiles across multiple tissue types.
Methodological approach: Comparative genomic analysis combined with functional assays in multiple model organisms can help delineate conserved versus divergent functions.
Multiple lines of evidence demonstrate BCL7C's tumor suppressor function in ovarian cancer:
BCL7C is downregulated in human ovarian carcinomas
Underexpression correlates with poor prognosis
Ectopic BCL7C restrains proliferation and invasion of ovarian cancer cells
BCL7C depletion reduces apoptosis while promoting proliferation and invasion
Primary ovarian carcinomas with low BCL7C levels often show elevated expression of mutant p53 target genes
Methodological approach: To investigate tumor suppressor properties, researchers should employ gain- and loss-of-function experiments in relevant cancer models, coupled with phenotypic assays measuring proliferation, invasion, and apoptosis.
Underexpression of BCL7C is associated with unfavorable prognosis in ovarian cancer and several other human cancers, suggesting its potential utility as a prognostic biomarker across multiple cancer contexts .
Methodological approach: Kaplan-Meier survival analysis stratified by BCL7C expression levels, multivariate Cox regression analysis controlling for known prognostic factors, and meta-analysis across multiple cancer datasets are recommended approaches.
While BCL7C appears to function as a tumor suppressor in ovarian cancer, research on BCL7B suggests potentially oncogenic roles in some cancer contexts. BCL7B has been identified as having high diagnostic and prognostic value in pan-cancer analysis .
Methodological approach: Comparative functional studies with simultaneous manipulation of multiple family members, followed by comprehensive phenotypic and transcriptomic profiling.
BCL7C suppresses mutant p53-mediated gene transcription by directly binding to mutant p53. Knockdown of BCL7C enhances the expression of mutant p53 target genes in ovarian cancer cells. This mechanism appears central to BCL7C's tumor suppressor function .
Methodological approach: Protein-protein interaction studies (co-immunoprecipitation, proximity ligation assays), ChIP-seq to identify genomic binding sites, and reporter assays to quantify transcriptional effects.
Yes, current evidence indicates a p53-dependent mechanism. BCL7C abrogates mutant p53-induced cell proliferation and invasion but shows no impact on proliferation and invasion of cancer cells with depleted p53 or those harboring wild-type p53 .
Methodological approach: Isogenic cell line models with varying p53 status (wild-type, mutant, null) combined with BCL7C manipulation provide the most controlled experimental system to test this dependency.
While direct evidence for BCL7C epigenetic regulation is limited in the provided search results, research in the field of epigenetics suggests potential regulatory mechanisms including DNA methylation, histone modifications, and chromatin accessibility .
Methodological approach: Integrated epigenomic analysis combining DNA methylation profiling, ChIP-seq for histone marks, ATAC-seq for chromatin accessibility, and correlation with expression data.
For CRISPR-based functional studies of BCL7C:
Use guide RNA sequences designed to efficiently target the BCL7C gene with minimal off-target effects
Order at least two gRNA constructs per gene to increase success rates
Verify gRNA sequences against your target gene sequence before ordering
Consider the specific splice variants or exons you wish to target
Methodological approach: Careful gRNA design with off-target prediction algorithms, validation of editing efficiency, and phenotypic confirmation across multiple independent clones.
Based on BCL7C's known functions, researchers should consider:
Proliferation assays (BrdU incorporation, Ki-67 staining)
Invasion assays (transwell, 3D spheroid invasion)
Apoptosis assays (Annexin V, caspase activation)
Gene expression analysis of mutant p53 target genes
Methodological approach: Multi-parametric approach combining real-time monitoring systems with endpoint assays to capture the dynamic nature of these cellular processes.
This represents a significant challenge in BCL7C research, particularly given its role in regulating transcription through mutant p53.
Methodological approach: Acute versus chronic depletion/overexpression systems, rescue experiments, and temporal analysis of molecular changes can help distinguish primary from secondary effects.
Given that BCL7C interacts with mutant p53, which acts as a transcription factor, and considering the importance of chromatin remodeling in cancer (as seen with SMARCA4/BRG1 ), the chromatin context likely influences BCL7C function.
Methodological approach: Combining ChIP-seq for BCL7C and mutant p53 with ATAC-seq and histone modification mapping in the same cellular contexts can provide insights into how chromatin states affect BCL7C activity.
While not directly addressed for BCL7C in the search results, research on SMARCA4/BRG1 has shown connections between chromatin remodeling factors and replication stress response . Similar relationships might exist for BCL7C.
Methodological approach: DNA fiber analysis, γH2AX foci quantification, and cell synchronization studies in BCL7C-manipulated cells during replication stress conditions.
Post-translational modifications often regulate protein function, stability, and interactions, but specific information about BCL7C modifications is limited in current research.
Methodological approach: Mass spectrometry-based proteomics focused on identifying phosphorylation, ubiquitination, acetylation, and other modifications under various cellular conditions.
Given BCL7C's association with prognosis in multiple cancer types, particularly ovarian cancer, integration into prognostic models represents a valuable research direction.
Methodological approach: Development of multi-factor prognostic signatures incorporating BCL7C expression with other established biomarkers, validated in independent patient cohorts using appropriate statistical methods.
Potential therapeutic approaches could include:
Restoration of BCL7C expression or function in cancers with downregulation
Combinatorial approaches targeting both BCL7C and mutant p53 pathways
Identification of synthetic lethal interactions in BCL7C-deficient contexts
Methodological approach: High-throughput drug screening in isogenic cell line pairs differing only in BCL7C status, followed by validation in patient-derived models.
Incorporating BCL7C status in clinical trial design could enhance the identification of responsive patient subgroups, particularly in cancers where BCL7C has prognostic significance.
Methodological approach: Retrospective analysis of existing trial data stratified by BCL7C expression, followed by prospective trials with pre-specified BCL7C-based subgroup analyses.
Different model systems offer distinct advantages:
Model System | Advantages | Limitations | Best Applications |
---|---|---|---|
Cancer cell lines | Easy manipulation, homogeneous | May not reflect tumor heterogeneity | Mechanism studies, initial drug screening |
Patient-derived xenografts | Maintain tumor heterogeneity | Expensive, lack immune component | Preclinical drug testing, biomarker validation |
Genetically engineered mouse models | In vivo context with intact immune system | Time-consuming, species differences | Developmental studies, immune interactions |
Clinical samples | Direct human relevance | Limited experimental manipulation | Biomarker studies, expression correlation |
Methodological approach: Multi-model validation of key findings across different systems to ensure robustness and translational relevance.
Various techniques offer different insights into BCL7C biology:
Technology | Measures | Sensitivity | Spatial Information | Best Applications |
---|---|---|---|---|
RT-qPCR | mRNA expression | High | None | Expression quantification |
RNAscope | mRNA expression | High | Cellular | Spatial expression patterns |
Western blot | Protein levels | Moderate | None | Protein expression, modifications |
IHC/IF | Protein levels | Moderate | Cellular/subcellular | Tissue localization |
Mass spectrometry | Protein, modifications | Variable | None | PTM identification |
Methodological approach: Integration of complementary techniques to build a comprehensive view of BCL7C biology at multiple levels.
Multi-omics integration is essential for comprehensive understanding of BCL7C function.
Methodological approach: Network-based integration of transcriptomic, proteomic, and epigenomic data using algorithms designed to identify causal relationships rather than mere correlations.
Key knowledge gaps include:
Comprehensive normal tissue expression patterns
BCL7C regulation at transcriptional and post-transcriptional levels
Functions beyond p53 interaction
Role in non-ovarian cancers
Potential as a therapeutic target
Methodological approach: Systematic literature review and meta-analysis to identify inconsistencies and unexplored areas, followed by targeted experimental approaches.
Single-cell approaches could reveal heterogeneity in BCL7C expression and function within tumors that is missed by bulk analysis.
Methodological approach: Single-cell RNA-seq, CyTOF, and spatial transcriptomics in tumor samples to map BCL7C expression patterns in relation to cell states and microenvironmental features.