BCL2L10 contains conserved Bcl-2 homology (BH) domains (BH1, BH2, and BH4) but lacks a canonical BH3 domain, contributing to its functional ambiguity . Key features include:
Anti-apoptotic activity: Binds BAX, BCL2, and BCL-XL to inhibit cytochrome c release and caspase activation .
Pro-apoptotic potential: Interacts with Apaf-1 and Caspase 9 in certain contexts, promoting apoptosis .
Non-apoptotic roles: Regulates cytoskeletal dynamics via Aurora kinase A (AURKA) interactions and metabolic pathways like the TCA cycle .
BCL2L10 exhibits tissue-specific oncogenic or tumor-suppressive functions:
Ovarian cancer: Knockdown induces G0/G1 cell cycle arrest, upregulates proliferation, and reduces expression of TCA cycle enzymes (SDHD, IDH1) .
Gastric cancer: Hypermethylation of BCL2L10 correlates with poor prognosis .
BCL2L10 modulates the TCA cycle in ovarian cancer:
Protein Target | Function | Impact of BCL2L10 Knockdown |
---|---|---|
SDHD | Succinate dehydrogenase subunit | ↓ Expression |
IDH1 | Isocitrate dehydrogenase 1 | ↓ Expression |
FH | Fumarate hydratase | No change |
This metabolic shift promotes glycolysis and proliferation, independent of apoptosis .
BCL2L10 forms complexes with key apoptotic regulators:
Targeting BCL2L10 in melanoma: Overexpression confers resistance to BRAF inhibitors (e.g., PLX-4032) and cisplatin .
Ovarian cancer: Downregulation correlates with metabolic dysregulation, suggesting potential for TCA cycle-targeted therapies .
BH3 mimetics: ABT-737 (a pan-BCL2 inhibitor) shows reduced efficacy in BCL2L10-overexpressing cells .
BH3 domain functionality: Conflicting reports on its presence/absence impact mechanistic models .
Tissue-specific effects: Opposite roles in ovarian vs. melanoma cancers remain unexplained .
Knockout phenotypes: Mouse Bcl2l10 knockouts show no developmental defects, complicating translational relevance .
BCL2L10 contains BH1, BH2, and BH4 domains as well as a putative carboxy-terminal transmembrane domain characteristic of anti-apoptotic Bcl-2 family proteins. While some studies report the presence of a pro-apoptotic BH3 domain in BCL2L10, conflicting evidence indicates an incomplete or absent BH3 domain . This structural ambiguity distinguishes BCL2L10 from other Bcl-2 family members and may explain its context-dependent functions.
Methodologically, researchers investigating BCL2L10 structure should employ both sequence analysis and experimental techniques such as site-directed mutagenesis to characterize functional domains. Western blotting can detect BCL2L10 at approximately 23 kDa using rabbit polyclonal antibodies .
Unlike the mouse homolog (Diva/Boo) which shows restricted expression primarily in adult ovary and testis, human BCL2L10 (Bcl-B) appears to be more widely expressed across tissues . This differential expression pattern between species suggests distinct evolutionary roles.
For studying tissue distribution, researchers should employ:
Quantitative RT-PCR for mRNA detection
Immunohistochemistry with validated antibodies for protein localization
Single-cell RNA sequencing for cell-type specific expression patterns
BCL2L10 functions by differentially binding other Bcl-2 family members and through interaction with the apoptosome protein Apaf-1 . These interactions determine whether BCL2L10 will exhibit pro-apoptotic or anti-apoptotic activity in a given cellular context.
To study these interactions, researchers should utilize:
Co-immunoprecipitation assays
Proximity ligation assays
FRET-based techniques for live-cell interaction studies
Molecular modeling based on known Bcl-2 family protein structures
For accurate BCL2L10 expression analysis, researchers should employ multiple complementary techniques:
When reporting expression data, researchers should normalize BCL2L10 levels to appropriate housekeeping genes or proteins and include statistical analysis of biological replicates.
Based on published methodologies, several approaches can effectively manipulate BCL2L10 expression:
RNA interference:
CRISPR-Cas9 gene editing:
Design guide RNAs targeting early exons
Confirm knockout via sequencing and protein detection
Overexpression systems:
Transfect cells with vectors containing BCL2L10 cDNA
Use inducible expression systems to control expression timing
Pharmacological modulation:
For validation, always include appropriate controls and analyze effects on related Bcl-2 family members to ensure specificity .
Select experimental models based on your specific research questions:
Cell line models:
Animal models:
Clinical samples:
Fresh or FFPE tumor samples with matched normal tissues
Organize by cancer subtype and clinical parameters
BCL2L10 promoter hypermethylation has been observed in gastric carcinoma, correlating significantly with decreased expression levels . This epigenetic silencing mechanism suggests BCL2L10 may function as a tumor suppressor in this cancer type.
When investigating promoter methylation:
Use bisulfite sequencing to map specific CpG sites affected
Correlate methylation patterns with expression levels
Treat cells with demethylating agents (e.g., 5-azacytidine) to confirm causality
Compare methylation patterns across cancer types to identify tissue-specific regulation
Research indicates that restoring BCL2L10 expression in hypermethylated gastric cancer cells induces apoptosis through mitochondrial pathways, supporting its tumor suppressor role in this context .
BCL2L10 is abundantly expressed in melanoma through STAT3-mediated transcriptional activation . Using reporter assays, site-directed mutagenesis, and ChIP analysis, researchers have identified functional STAT3 responsive elements in the BCL2L10 promoter.
To investigate this regulatory relationship:
Examine correlation between phosphorylated STAT3 and BCL2L10 expression
Treat melanoma cells with STAT3 inhibitors and measure BCL2L10 expression changes
Perform promoter mutation studies to confirm STAT3 binding sites
Analyze clinical samples for co-expression patterns
In melanoma, BCL2L10 acts as a pro-survival factor, protecting cells from cytotoxic effects of various drugs including cisplatin, dacarbazine, and ABT-737, as well as combination treatments with BRAF inhibitors .
The contradictory functions of BCL2L10 across different cancers represent a fascinating research area:
To reconcile these contradictory roles, researchers should:
Conduct comprehensive protein interaction studies in different cellular contexts
Investigate tissue-specific post-translational modifications
Examine potential isoform expression differences
Analyze the influence of the tumor microenvironment on BCL2L10 function
In melanoma, BCL2L10 functions as a pro-survival factor, protecting cells from the cytotoxic effects of different drugs, including cisplatin, dacarbazine, and ABT-737 . BCL2L10 also inhibits cell death upon combination treatments of PLX-4032 (a BRAF inhibitor) with ABT-737 or cisplatin.
For studying BCL2L10's role in drug response:
Compare apoptotic responses in BCL2L10-expressing versus depleted cells
Analyze activation of caspases and other apoptotic mediators
Measure mitochondrial membrane potential changes
Perform drug resistance profiling with dose-response curves
Understanding BCL2L10's role in drug resistance could inform combination therapy strategies that overcome its anti-apoptotic effects in cancers where it functions as an oncogene.
Studies in mouse models indicate BCL2L10 (Diva/Boo) plays a role in oocyte maturation . RNAi-mediated knockdown of Bcl2l10 in mouse germinal vesicle oocytes affects meiotic progression, with many oocytes arresting at metaphase I.
To investigate developmental roles:
Examine expression patterns during different developmental stages
Perform lineage-specific knockdown/knockout studies
Analyze effects on differentiation markers and cell fate decisions
Compare phenotypes across model organisms
Interestingly, despite its expression in mouse ovary and testis, Bcl2l10 knockout mice were fertile with no obvious developmental defects , suggesting possible compensatory mechanisms.
Although the search results don't specifically address post-translational modifications of BCL2L10, this represents an important research area that may explain its context-dependent functions.
Researchers investigating this aspect should:
Perform mass spectrometry to identify phosphorylation, ubiquitination, or other modifications
Create site-specific mutants to determine functional consequences
Identify enzymes responsible for these modifications
Examine modification patterns in different cellular contexts
Understanding post-translational regulation could reveal mechanisms by which BCL2L10 switches between pro- and anti-apoptotic functions in different tissues or disease states.
The prognostic significance of BCL2L10 appears to be cancer-type dependent:
In gastric carcinoma, low BCL2L10 expression due to promoter hypermethylation suggests it may serve as a tumor suppressor biomarker
In melanoma, high BCL2L10 expression correlates with resistance to various drugs, suggesting potential as a predictive biomarker for treatment response
For clinical correlation studies:
Use standardized scoring systems for immunohistochemistry
Correlate expression with survival outcomes using Kaplan-Meier analyses
Stratify patients by cancer subtype and treatment history
Perform multivariate analyses to determine independent prognostic value
Based on the dual nature of BCL2L10, therapeutic strategies should be cancer-type specific:
For cancers where BCL2L10 is oncogenic (e.g., melanoma):
Develop specific BCL2L10 inhibitors
Target upstream regulators like STAT3
Combine with existing BH3 mimetics like ABT-737
For cancers where BCL2L10 is suppressed (e.g., gastric cancer):
Use epigenetic modulators to reverse promoter methylation
Develop approaches to restore or mimic BCL2L10 function
Target pathways activated by BCL2L10 loss
When designing inhibitor screens, researchers should employ:
Structure-based drug design leveraging BCL2L10 protein structure
Cell-based phenotypic screens measuring apoptotic responses
In vivo validation in appropriate animal models
For clinical monitoring of BCL2L10:
Approach | Application | Advantages | Limitations |
---|---|---|---|
Immunohistochemistry | Tumor samples | Cellular localization, widely available | Semi-quantitative, interpreter variability |
qRT-PCR | Tissue or liquid biopsy | Quantitative, sensitive | No protein information |
Methylation analysis | Tissue or liquid biopsy | Detects epigenetic silencing | Cancer-type specific relevance |
Circulating tumor DNA | Liquid biopsy | Non-invasive monitoring | Technical challenges, standardization |
Development of companion diagnostics for BCL2L10 status would be particularly valuable for cancers where it influences treatment response, such as melanoma .
BCL2L10 contains several conserved domains, including the BH4, BH1, and BH2 domains . These domains are critical for its function and interactions with other proteins in the BCL-2 family. BCL2L10 can form hetero- or homodimers with other BCL-2 family members, influencing their pro- or anti-apoptotic activities .
The primary function of BCL2L10 is to suppress apoptosis. It achieves this by preventing the release of cytochrome c from the mitochondria, a key step in the activation of caspase-3, an enzyme that plays a central role in the execution phase of cell apoptosis . By inhibiting this pathway, BCL2L10 helps in promoting cell survival.
Recombinant BCL2L10 is produced using recombinant DNA technology, where the gene encoding BCL2L10 is cloned and expressed in a suitable host, such as E. coli. The recombinant protein is then purified using chromatographic techniques . The recombinant human BCL2L10 typically consists of 195 amino acids and has a molecular mass of approximately 21.8 kDa .
Recombinant BCL2L10 is used in various research applications, including: