Protein Identity:
Production:
Inhibits mitochondrial outer membrane permeabilization (MOMP) by sequestering Bax/Bak and BH3-only proteins (e.g., Bim, Puma) .
Modulates ER calcium homeostasis by interacting with IP3 receptors (IP3Rs), promoting oscillatory calcium release linked to cell survival .
Suppresses autophagy by binding Beclin-1 via its BH3-like motif .
Therapeutic Targeting:
Disease Models:
Stem Cell Biology:
Human Bcl-2 (1-206 a.a.) is a 26 kDa protein containing four Bcl-2 homology domains (BH1-BH4) that form a hydrophobic groove capable of binding BH3 domains of pro-apoptotic proteins. The protein functions primarily by sequestering BH3-only proteins, preventing them from activating the pro-apoptotic executioner proteins BAX and BAK .
Bcl-2 expression is dynamically regulated during cellular differentiation, as evidenced in dendritic cells (DCs) where expression decreases during maturation. Research shows that immature CD11c+MHC-II(I-A)+ DCs express significantly higher Bcl-2 levels than mature CD11c+MHC-II(I-A)++ DCs .
To accurately detect these changes, researchers should:
Use intracellular staining with specific anti-Bcl-2 antibodies followed by flow cytometry
Calculate the ratio between mean fluorescence intensities (MFI) of Bcl-2 staining and isotype control staining to correct for background signals
Include positive control populations (such as CD8+ T cells that express high Bcl-2 levels) within samples to validate staining efficacy
Complement flow cytometry data with Western blot or RT-qPCR analysis
Bcl-2 demonstrates distinctive binding preferences among pro-survival family members (including Bcl-xL, Bcl-w, Mcl-1, Bfl-1/A1, and Bcl-B). These differences stem from subtle variations in the BH3-binding groove structure:
Pro-survival Protein | Preferential Binding Partners | Binding Affinity Range |
---|---|---|
Bcl-2 | BIM, BAD, BIK | Picomolar to nanomolar |
Bcl-xL | BIM, BID, BAD, BIK | Picomolar to nanomolar |
Mcl-1 | BIM, NOXA, PUMA | Nanomolar |
Bfl-1/A1 | BIM, NOXA, PUMA | Nanomolar |
Bcl-w | BIM, BAD | Nanomolar |
Bcl-B | BIM (weakly) | Micromolar |
These binding specificities create a complex interaction network that determines cellular susceptibility to apoptosis . Understanding these distinctions is crucial for designing specific inhibitors and predicting therapeutic responses.
For successful expression and purification of human Bcl-2 (1-206 a.a.), researchers should consider:
Expression system selection:
E. coli expression with metal affinity tags (His-tag) provides high yield but may require refolding
Expression in insect cells may improve solubility but with lower yields
Optimal protocol:
Clone the coding sequence into a vector with an N-terminal His-tag
Express in E. coli BL21(DE3) strain at lower temperatures (16-18°C)
Purify using metal affinity chromatography followed by gel filtration
Consider truncating the C-terminal transmembrane domain (amino acids 207-239) to improve solubility
Common challenges and solutions:
Protein aggregation: Add mild detergents (0.1% CHAPS) to purification buffers
Poor yield: Optimize codon usage for E. coli expression
Loss of function: Verify proper folding using circular dichroism and thermal shift assays
Stability during storage: Add 10% glycerol and store at -80°C in small aliquots
Several complementary techniques offer robust analysis of Bcl-2 protein interactions:
Bio-layer interferometry (BLI):
X-ray crystallography:
Cross-linking coupled with mass spectrometry:
Yeast surface display:
Fluorescence resonance energy transfer (FRET):
Enables detection of interactions in living cells
Provides spatial information about protein proximities
Requires fluorescent protein tagging that may affect native interactions
To systematically determine BCL2 dependency profiles in cancer cell lines:
Combinatorial inhibition strategy:
Recommended methodology:
Treat cells with dose ranges of specific inhibitors (BH3-mimetics or designed protein inhibitors)
Assess viability using multiple assays (e.g., MTT, CellTiter-Glo)
Measure apoptosis markers (Annexin V/PI staining, caspase activation)
Confirm findings using genetic approaches (CRISPR knockout or RNAi)
Data analysis approach:
Validation experiments:
Interpreting discrepancies between Bcl-2 expression and clinical outcomes requires consideration of several factors:
Context-dependent function:
Methodological considerations:
Integrated interpretation:
Functional validation:
To identify and overcome resistance to Bcl-2 inhibitors:
Resistance mechanism identification:
Overcoming primary resistance:
Addressing acquired resistance:
Translational considerations:
Differential Bcl-2 expression patterns across cancer subtypes significantly impact therapeutic approaches:
Hematologic malignancies:
Solid tumors:
Different dependencies identified through combinatorial inhibition:
Predictive biomarkers approach:
Toxicity management strategies:
Developing highly specific Bcl-2 inhibitors requires sophisticated computational and structural approaches:
De novo protein design strategy:
Structure-guided optimization:
Crystal structures of inhibitor-target complexes (e.g., αMCL1- Mcl-1 at 2.75 Å resolution) provide detailed binding information
Analysis of binding pockets reveals subtle differences that can be exploited for specificity
Precise positioning of designed sidechains enables high affinity and selectivity
Directed evolution approaches:
Validation and refinement:
To distinguish between canonical and non-canonical Bcl-2 functions:
Domain-specific mutants approach:
Subcellular localization studies:
Create Bcl-2 variants with specific localization signals (mitochondrial, ER, nuclear)
Compare phenotypic effects of differentially localized variants
Use microscopy and fractionation to confirm localization
Temporal dynamics analysis:
Use inducible expression systems to distinguish immediate versus delayed effects
Implement rapid protein degradation systems (e.g., AID, dTAG) for acute depletion
Compare acute versus chronic inhibition phenotypes
Interactome profiling:
Multi-omics integration:
To study Bcl-2's role in immune regulation, researchers should consider:
Transgenic mouse models:
Flow cytometry analysis approach:
Functional immunological assays:
In vivo immunization models:
Single-cell approaches:
scRNA-seq to capture heterogeneity within immune populations
Trajectory analysis to identify developmental stages where Bcl-2 is critical
Spatial transcriptomics to understand Bcl-2 regulation in tissue microenvironments
Several cutting-edge approaches show promise for overcoming current limitations:
Proteolysis targeting chimeras (PROTACs):
Antibody-drug conjugates (ADCs):
BH4 domain-targeting approaches:
Spatiotemporally controlled targeting:
RNA-targeting therapies:
Antisense oligonucleotides
siRNA delivery systems
mRNA destabilizing approaches
To better understand Bcl-2 family interplay:
Multiplexed genetic manipulation:
CRISPR-based screens targeting multiple family members simultaneously
Inducible expression systems with orthogonal control
Base editing to introduce specific mutations rather than knockouts
Proximity-based interaction mapping:
BioID or APEX2 proximity labeling to identify spatial interaction networks
Split-protein complementation assays to visualize interactions in living cells
Advanced FRET/BRET systems with improved dynamic range
Dynamic measurement systems:
Live-cell reporters of apoptotic pathway activation
Optogenetic control of individual protein activities
Biosensors to monitor conformational changes in real-time
Patient-derived experimental models:
Organoids from primary tumors
Patient-derived xenografts
Ex vivo culture systems for primary cells
Systems biology approaches:
Key methodological challenges and approaches include:
Site-specific modification detection:
Develop specific antibodies against common Bcl-2 modifications (phosphorylation, ubiquitination)
Employ targeted mass spectrometry approaches (multiple reaction monitoring)
Use biochemical enrichment strategies for low-abundance modified forms
Modification dynamics analysis:
Pulse-chase approaches to determine modification turnover rates
Single-cell techniques to capture cell-to-cell variability
Time-resolved proteomics following cellular stimulation
Functional impact assessment:
Generate modification-mimetic mutants (e.g., phosphomimetic S→D/E substitutions)
Create modification-resistant mutants (e.g., S→A substitutions)
Develop conformation-specific antibodies that detect functionally distinct states
Structural consequences:
Use hydrogen-deuterium exchange mass spectrometry to detect conformational changes
NMR studies of modified versus unmodified proteins
Molecular dynamics simulations to predict modification effects
Spatiotemporal regulation:
Develop biosensors to track modifications in live cells
High-resolution microscopy to detect subcellular localization changes
Correlate modifications with protein-protein interaction dynamics
B-Cell Leukemia/Lymphoma 2 (BCL-2) is a protein encoded by the BCL2 gene in humans. It is a member of the BCL-2 family of regulator proteins that regulate cell death (apoptosis) by either inducing or inhibiting apoptosis. The BCL-2 protein is crucial in the regulation of the mitochondrial pathway of apoptosis, which is a key mechanism in the maintenance of cellular homeostasis and the prevention of cancer.
The BCL-2 protein consists of several domains, including the BH1, BH2, BH3, and BH4 domains, which are involved in its anti-apoptotic function. The (1-206 a.a.) fragment of the BCL-2 protein represents the first 206 amino acids of the protein, which includes the BH4 domain and part of the BH3 domain. This fragment is significant because it retains the ability to inhibit apoptosis, making it a valuable tool for research and therapeutic applications.
BCL-2 is known for its role in cancer, particularly in B-cell malignancies such as B-cell leukemia and lymphoma. Overexpression of BCL-2 has been observed in various cancers, where it contributes to the resistance of cancer cells to apoptosis, allowing them to survive and proliferate uncontrollably. This makes BCL-2 a target for cancer therapy, with several BCL-2 inhibitors being developed and tested in clinical trials.
The human recombinant BCL-2 (1-206 a.a.) is a laboratory-produced version of the BCL-2 protein fragment. It is used in research to study the function of BCL-2 and its role in apoptosis and cancer. Recombinant proteins are produced using recombinant DNA technology, which involves inserting the gene encoding the protein into a host cell, such as bacteria or yeast, which then produces the protein.
The recombinant BCL-2 (1-206 a.a.) protein is used in various research applications, including: