The recombinant protein is synthesized using optimized E. coli expression systems and purified via proprietary chromatographic techniques ( ).
Anti-Apoptotic Activity: Binds pro-apoptotic proteins (e.g., Bax, Bak) and prevents cytochrome c release from mitochondria, inhibiting caspase activation ( ).
Homodimerization: Cytosolic Bcl-xL forms homodimers via C-terminal tail interactions, a mechanism critical for mitochondrial membrane insertion and activity ( ).
Role in Erythropoiesis: Essential for survival of human erythroid progenitors and hematopoietic stem cells ( ).
Therapeutic Target: Bcl-xL overexpression in cancers promotes chemoresistance. Inhibitors like WEHI-539 and Navitoclax are under investigation ( ).
Metastasis: Nuclear-localized Bcl-xL drives epithelial–mesenchymal transition (EMT) and metastasis independently of its anti-apoptotic function ( ).
Pancreatic Progenitors: Bcl-xL supports survival of differentiating pancreatic cells from human pluripotent stem cells (hPSCs). Inhibition reduces PDX1 and HNF4A expression, impairing beta-cell maturation ( ).
BCL-XL (BCL2L1) is an essential anti-apoptotic protein belonging to the BCL-2 family, which includes BCL-2, MCL-1, A1, and BCL-W. These proteins counteract apoptotic signals that emerge during development and under stress conditions. BCL-XL was the first identified BCL-2 homologue and represents one of two splicing variants of the BCL-X gene. It functions by binding to pro-apoptotic proteins such as BIM, BMF, BAD, BIK, HRK, PUMA, tBID, and also to BAX and BAK. By shuttling BAX from mitochondria to cytosol, BCL-XL reduces BAX levels at mitochondria and decreases cellular apoptotic susceptibility .
Recombinant His-tagged BCL-XL typically refers to E. coli-derived human BCL-XL protein (often Ser2-Arg212) with a C-terminal 6-His tag added for purification purposes. This tag allows for efficient isolation using metal affinity chromatography without significantly affecting its biological function. The recombinant protein may also lack the C-terminal transmembrane domain (minus C-terminus) to improve solubility. While functionally similar to native BCL-XL in binding assays, researchers should verify that the His-tag doesn't interfere with specific experimental applications, particularly those involving membrane interactions or structural studies .
For optimal stability and activity, His-tagged BCL-XL protein should be stored in a manual defrost freezer, avoiding repeated freeze-thaw cycles. When shipped, the protein is typically supplied as a 0.2 μm filtered solution in HEPES and KCl and transported with dry ice or equivalent cooling. Upon receipt, it should be immediately stored at the recommended temperature. For experimental use, carrier-free versions (without BSA) are recommended for applications where BSA might interfere, while versions with carrier protein provide enhanced stability and longer shelf-life for general applications like cell culture or as ELISA standards .
To conduct cytochrome c release assays for BCL-XL inhibition studies, researchers should implement the following methodology:
Prepare or obtain crude/enriched mouse liver mitochondria following established protocols
Formulate appropriate buffers:
Dilution Buffer: 25 mM HEPES-KOH (pH 7.4), 0.1 M KCl, 10% Glycerol, 1 mg/mL fatty acid-free BSA
Mitochondria Buffer: 125 mM KCl, 0.5 mM MgCl2, 3.0 mM Succinic acid, 3.0 mM Glutamic acid, 10 mM HEPES-KOH (pH 7.4), 1 mg/mL BSA, with protease inhibitors (25 μg/mL Leupeptin, 25 μg/mL Pepstatin, 3 μg/mL Aprotinin, 100 μM PMSF, and 10 μM caspase inhibitor)
Use recombinant Human BCL-XL (minus C-Terminus) protein with optimization at concentrations <250 nM
Include recombinant Human BID Caspase-8-cleaved (54 nM) as a positive control
Measure cytochrome c release through appropriate detection methods such as Western blotting or ELISA
Laboratories should determine optimal dilutions for their specific experimental conditions .
For effective BCL-XL knockdown studies in human hematopoietic cells, researchers should:
Use lentiviral vectors expressing shRNAs targeting multiple regions of BCL-XL mRNA to ensure efficient knockdown
Include appropriate controls:
Non-targeting shRNA control
Rescue experiments with BCL-XL overexpression constructs resistant to the shRNA
BCL-2 overexpression controls (as BCL-2 can compensate for BCL-XL deficiency)
Assess knockdown efficiency through both mRNA (qRT-PCR) and protein (Western blot) analyses
Evaluate effects at multiple stages of hematopoiesis, from early stem cells to mature blood cells
Compare genetic knockdown effects with chemical inhibition using BH3-mimetics to validate findings
For functional assessments, include colony formation assays, apoptosis measurements, and in vivo xenotransplantation models
This approach has revealed that BCL-XL inhibition affects human hematopoietic cells more profoundly than anticipated from murine studies, with significant losses observed from early erythropoiesis stages and reduced megakaryocyte populations .
When designing Proteolysis-Targeting Chimeras (PROTACs) directed at BCL-XL, researchers should consider these critical factors:
Lysine accessibility: Computational modeling of the entire NEDD8-VHL Cullin RING E3 ubiquitin ligase complex with BCL-XL reveals that only lysines in specific band regions can be effectively ubiquitinated, directly affecting degrader potency
E3 ligase selection: VHL-recruiting PROTACs have successfully generated potent BCL-XL/BCL-2 dual degraders with improved antitumor activity against dependent leukemia cells
Warhead optimization: ABT263-derived binding moieties provide effective starting points for recognition of the BH3 binding groove
Linker design: The length, composition, and attachment point critically influence the positioning of BCL-XL relative to the E3 ligase complex, determining which lysines become accessible for ubiquitination
Selectivity profile: Determine whether selective BCL-XL degradation or dual BCL-XL/BCL-2 targeting provides optimal therapeutic outcomes for specific indications
This structure-guided approach represents a conceptual framework for developing PROTACs with improved selectivity and potency .
BCL-XL dependency in human erythropoiesis is far more extensive than previously understood from mouse models. While murine studies indicated BCL-XL importance primarily in late erythroid stages, human research reveals:
BCL-XL is critical from the earliest stages of erythropoiesis, with efficient genetic or chemical inhibition causing significant loss of early human erythroid progenitors
This dependency continues throughout erythroid differentiation, making the entire red blood cell production lineage vulnerable to BCL-XL inhibition
The magnitude of BCL-XL dependency in human erythroid cells exceeds what would be predicted from murine data or early clinical trials with BCL-XL inhibitors
BCL-XL deficiency in human erythroid precursors is not rescued by loss of its antagonist BIM, suggesting involvement of other pro-apoptotic factors
BCL-2 overexpression can fully compensate for BCL-XL deficiency in erythroid cells, indicating potential functional redundancy between these proteins
These findings have significant implications for developing BCL-XL inhibitors as therapeutics, predicting more severe anemia than observed in early clinical trials with pan-BCL-2 family inhibitors .
BCL-XL inhibition profoundly affects human hematopoietic stem cells (HSCs) and multipotent progenitors in ways not predicted by mouse studies:
BCL-XL deficient human HSCs and progenitors show significant numerical reduction
These cells demonstrate severely impaired capacity to engraft during xenotransplantation experiments in mice
This contrasts with murine studies where Bcl-x deletion in adult hematopoietic cells did not noticeably affect the HSPC compartment
BCL-XL deficiency in human HSCs can be fully compensated by BCL-2 overexpression
Loss of the pro-apoptotic protein BIM does not rescue human HSCs from BCL-XL deficiency
These findings suggest that specific BCL-XL inhibitors may cause more severe hematological toxicities than previously anticipated, potentially affecting not only mature blood cells but also the stem cell compartment .
Megakaryocytes and platelets show significant BCL-XL dependency through several mechanisms:
Clinical studies with Navitoclax (ABT-263), a combined BCL-2/BCL-XL/BCL-W inhibitor, demonstrate severe thrombocytopenia caused by direct platelet destruction
This thrombocytopenia is partially counteracted by increased megakaryopoiesis, suggesting a compensatory response
Genetic BCL-XL inhibition studies confirm reduction in megakaryocyte populations
Platelets are particularly vulnerable to BCL-XL inhibition due to their inability to synthesize new proteins and limited alternative survival mechanisms
The platelet dependency on BCL-XL represents a primary dose-limiting toxicity for BCL-XL-targeting therapeutics
These findings explain the clinical observation of thrombocytopenia with BCL-XL inhibitors and highlight the challenge of developing such agents with acceptable therapeutic windows .
The significant discrepancies between mouse and human BCL-XL dependency patterns require careful analysis:
Experimental approaches:
Conduct direct comparative studies using identical methodologies
Perform comprehensive expression profiling of all BCL-2 family members across species and cell types
Utilize humanized mouse models to better recapitulate human hematopoiesis
Mechanistic considerations:
Examine species-specific differences in expression patterns of other anti-apoptotic proteins that might compensate for BCL-XL in mice but not humans
Investigate variations in mitochondrial priming states between species
Analyze differences in upstream regulatory pathways controlling BCL-XL function
Interpretation guidelines:
Exercise caution when extrapolating murine findings to human therapeutic applications
Prioritize data from human primary cells and humanized models for clinical development decisions
Consider species differences when establishing dosing strategies and toxicity monitoring
These reconciliation approaches are essential for accurate prediction of therapeutic outcomes and side effect profiles when targeting BCL-XL in humans .
The differential impact of BCL-XL deficiency on human versus mouse HSCs involves several potential molecular mechanisms:
Expression profile differences:
Human HSCs may have different basal levels of pro-apoptotic BCL-2 family members
The compensatory capacity of other anti-apoptotic proteins likely differs between species
Cellular stress handling:
Human and mouse HSCs may experience different levels of metabolic or replicative stress
Species-specific differences in DNA damage response pathways may contribute
Mitochondrial dynamics:
Human HSCs may have different mitochondrial priming states (proximity to apoptotic threshold)
Species-specific differences in mitochondrial structure or function could influence BCL-XL dependency
Microenvironmental factors:
Differences in niche interactions between human and mouse HSCs may affect their reliance on anti-apoptotic proteins
Cytokine responsiveness varies between species, potentially altering survival requirements
The finding that BCL-2 overexpression can rescue BCL-XL deficiency, while BIM deletion cannot, suggests complex mechanisms involving multiple proteins beyond a simple BCL-XL/BIM axis .
Lysine accessibility plays a critical role in determining the efficacy of PROTAC-mediated BCL-XL degradation:
Based on the established role of BCL-XL in human hematopoiesis, several hematological malignancies emerge as potential candidates for BCL-XL-targeting therapeutics:
Polycythemia vera: This myeloproliferative neoplasm characterized by excessive red blood cell production may be sensitive to BCL-XL inhibition given the critical dependency of the erythroid lineage on BCL-XL
Acute erythroid leukemia: Malignant transformation of erythroid precursors likely preserves or enhances BCL-XL dependency, potentially making these rare leukemias particularly vulnerable
Essential thrombocytosis: Although platelets are highly BCL-XL dependent, malignant megakaryocytes driving excessive platelet production may represent therapeutic targets if selective delivery can be achieved
Acute megakaryocytic leukemia: The megakaryocytic origin of these leukemias suggests potential sensitivity to BCL-XL inhibition
BCL-XL-dependent lymphoid malignancies: Certain lymphomas and leukemias may exhibit BCL-XL dependency despite originating from lineages where BCL-2 typically predominates
Therapeutic success will require balancing efficacy against the expected hematological toxicities, particularly thrombocytopenia and anemia .
To effectively assess and predict hematological toxicities of novel BCL-XL inhibitors, researchers should implement a comprehensive evaluation strategy:
In vitro assessment:
Test effects on primary human HSCs, erythroid precursors, and megakaryocytes at multiple differentiation stages
Compare with established inhibitors like Navitoclax to benchmark toxicity profiles
Evaluate concentration-dependent effects to establish therapeutic windows
Ex vivo systems:
Utilize whole blood assays to assess platelet survival
Implement colony formation assays with primary human bone marrow
Develop organoid or 3D culture systems that better recapitulate bone marrow niches
In vivo models:
Employ humanized mouse models with human hematopoietic system reconstitution
Monitor multiple hematological parameters including stem cell function
Assess long-term effects on hematopoietic recovery
Biomarker development:
Identify early indicators of hematological toxicity
Establish correlation between biomarker changes and functional hematological outcomes
These approaches will provide more accurate predictions of human toxicities than conventional mouse models alone, given the demonstrated species differences in BCL-XL dependency patterns .
Several innovative approaches may enhance the therapeutic window of BCL-XL-targeting compounds:
Targeted delivery systems:
Antibody-drug conjugates targeting malignancy-specific antigens
Nanoparticle formulations with preferential tumor accumulation
Tumor microenvironment-activated prodrugs
Advanced drug design:
Dual BCL-XL/BCL-2 inhibitors optimized for specific malignancies
Development of PROTACs that achieve catalytic degradation, potentially requiring lower drug exposure
Tissue-selective E3 ligase recruiting moieties to direct degradation preferentially in malignant cells
Combination strategies:
Pairing with agents that selectively sensitize malignant cells
Identifying synergistic combinations allowing lower BCL-XL inhibitor doses
Temporary cytoprotection of normal hematopoietic cells during treatment
Modified administration approaches:
Optimized dosing schedules allowing hematological recovery between treatments
Pulsatile high-concentration exposure rather than continuous dosing
Pre-emptive growth factor support to counteract specific toxicities
These strategies aim to maintain therapeutic efficacy while minimizing the impact on normal hematopoietic cells that depend on BCL-XL for survival .
B-Cell Lymphoma Extra Large (Bcl-XL) is a protein that plays a crucial role in the regulation of apoptosis, or programmed cell death. It is a member of the Bcl-2 family of proteins, which are known for their ability to regulate cell death by either promoting or inhibiting apoptosis. The human recombinant form of Bcl-XL, tagged with a His (histidine) tag, is commonly used in research to study its structure and function.
Bcl-XL is an anti-apoptotic protein that helps in the survival of cells by inhibiting the apoptotic pathways. It is involved in the formation of heterodimers with pro-apoptotic proteins, which is a significant event in the regulation of apoptosis . The His tag is a sequence of histidine residues added to the protein to facilitate its purification and detection in laboratory experiments.
Bcl-XL is particularly important in the context of cancer research. It is known to be involved in the survival of cancer cells, making it a target for cancer therapy . By inhibiting apoptosis, Bcl-XL allows cancer cells to evade the natural cell death process, leading to uncontrolled cell proliferation.
The human recombinant form of Bcl-XL is produced in E. coli and is a single, non-glycosylated polypeptide chain containing amino acids 1-210 . The His tag allows for easy purification using affinity chromatography techniques. This recombinant protein is used in various research applications to study its role in apoptosis and cancer.
Researchers use the human recombinant Bcl-XL protein to understand its structure, function, and interactions with other proteins. It is also used in drug discovery to identify potential inhibitors that can block its anti-apoptotic function, thereby promoting the death of cancer cells.