Bcl XL Human

B-Cell Lymphoma Extra Large Human Recombinant
Shipped with Ice Packs
In Stock

Description

Anti-Apoptotic Mechanism

Bcl-xL prevents mitochondrial outer membrane permeabilization (MOMP) by:

  • Binding pro-apoptotic proteins (BIM, BAD, BAX, BAK) to block their activation .

  • Sequestrating BAX/BAK at mitochondria, reducing cytochrome c release .

  • Essential for embryogenesis and hematopoietic cell survival, as shown in knockout models .

Non-Canonical Roles in Cancer Metastasis

Recent studies reveal metastasis-promoting functions independent of apoptosis inhibition:

Nuclear Localization and Epigenetic Regulation

  • Nuclear Bcl-xL (not mitochondrial) induces epithelial–mesenchymal transition (EMT) and stemness in pancreatic neuroendocrine tumors (panNETs) and breast cancer .

  • Mechanism: Recruits histone H3K4 trimethylation (H3K4me3) to the TGFβ promoter, enhancing TGFβ signaling .

    • TGFβ upregulation promotes autocrine/paracrine effects, including immunosuppression and extracellular matrix degradation .

Metastatic Potential in vivo

  • Apoptosis-defective Bcl-xL mutants (e.g., Mutant 1 with nuclear localization) drive metastasis in xenograft models .

  • Clinical correlation: Nuclear Bcl-xL is detected in 70% of metastatic panNETs vs. mitochondrial localization in primary tumors .

Table 2: Metastatic Functions of Bcl-xL Variants

VariantLocalizationApoptosis InhibitionMetastatic ActivityReference
Wild-type Bcl-xLMitochondriaYesModerate
Mutant 1 (ΔBH4)NucleusNoHigh
Mutant 2 (ΔTM)CytosolNoLow

Engineered Bcl-xL Constructs

Plasmids encoding subcellular-targeted Bcl-xL variants (Kerafast) :

  • Wild-type: Mitochondrial localization.

  • ΔC-terminal: Cytosolic.

  • ER-targeted: Engineered for endoplasmic reticulum retention.

Inhibitors and Therapeutics

  • WEHI-539 and A-1331852: Selective Bcl-xL inhibitors shown to reduce tumor-associated neutrophil survival in lung adenocarcinoma models .

  • Venetoclax combinations: Bcl-xL inhibition enhances efficacy in chemotherapy-resistant cancers .

Clinical and Therapeutic Implications

  • Cancer: Overexpression correlates with poor prognosis in panNETs, breast cancer, and leukemia .

  • Neutrophil targeting: Bcl-xL inhibition selectively eliminates pro-tumor neutrophils without affecting peripheral populations .

  • Challenges: Tissue-specific toxicity (e.g., thrombocytopenia) limits systemic Bcl-xL inhibition .

Product Specs

Introduction
Bcl-XL, a transmembrane protein residing in the mitochondrial membranes of long-lived and postmitotic cells like adult brain cells, participates in the FAS-Ligand signal transduction pathway. As an anti-apoptotic protein within the Bcl-2 family, Bcl-XL forms heterodimers, playing a crucial role in apoptosis regulation. Notably, BCL-XL contributes to cancer cell survival. Its primary function involves monitoring and suppressing apoptosis, effectively acting as a pro-survival protein.
Description
Recombinant human Bcl-XL, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising amino acids 1 to 210. The purification of Bcl-XL is achieved through proprietary chromatographic techniques.
Physical Appearance
White lyophilized (freeze-dried) powder, sterile filtered.
Formulation
The protein is supplied in a buffer containing 10mM Tris-HCl (pH 8), 1mM EDTA, and 250mM NaCl.
Solubility
To reconstitute, suspend Bcl-XL in 1000µl of sterile, 18M-cm H2O and allow it to dissolve overnight at 4°C. Dilute the reconstituted protein 10-fold into the desired buffer system. Due to Bcl-XL's propensity to form intramolecular disulfide bonds, the addition of 5mM DTT to the assay buffer is recommended. For SDS-PAGE analysis, 10mM DTT is advised.
Stability
Lyophilized Bcl-XL remains stable at room temperature for up to 3 weeks; however, it is recommended to store the desiccated product below -18°C. After reconstitution, Bcl-XL should be stored at 4°C for 2-7 days. For long-term storage, aliquot and freeze at -18°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of Bcl-XL is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
BclXL, Bcl-X(L), Bcl-XL.
Source
Escherichia Coli.

Q&A

What is BCL-XL and what is its primary function in human cells?

BCL-XL (B-cell lymphoma-extra large) is an anti-apoptotic protein belonging to the BCL-2 family that localizes primarily to the mitochondria. It functions as a key regulator of the intrinsic apoptotic pathway by preventing mitochondrial outer membrane permeabilization. BCL-XL exerts its anti-apoptotic activity by binding to and sequestering pro-apoptotic proteins such as BAX and BAK, thereby preventing cytochrome c release and subsequent caspase activation. This interaction is critical for maintaining cellular homeostasis and preventing inappropriate cell death under normal physiological conditions . Additionally, BCL-XL can interact with Beclin 1, a key regulator of autophagy, suggesting interconnected roles between apoptotic and autophagic pathways .

What techniques are most reliable for quantifying BCL-XL expression in human tissues?

For accurate quantification of BCL-XL expression in human tissues, researchers should consider a combination of techniques. Immunohistochemistry (IHC) provides spatial information but requires careful validation of antibody specificity and standardized scoring systems. Quantitative approaches like Western blotting with densitometry analysis offer more objective quantification when normalized to appropriate housekeeping proteins. RT-qPCR provides sensitive mRNA expression analysis, while RNA sequencing offers comprehensive transcriptional profiling. In bioinformatic analyses, as demonstrated in pancreatic ductal adenocarcinoma studies, BCL-XL expression can be reliably quantified from microarray data, showing excellent discrimination capacity (AUC: 0.83 [95% CI: 0.76, 0.90]; p < 0.001) between tumor and normal tissues . For clinical samples, tissue microarrays enable high-throughput analysis across multiple specimens with standardized staining conditions. Regardless of the technique chosen, inclusion of appropriate positive and negative controls is essential for reliable quantification.

How does BCL-XL promote metastasis independently of its anti-apoptotic function?

BCL-XL promotes metastasis through mechanisms distinct from its canonical anti-apoptotic activity at the mitochondria. Studies in pancreatic neuroendocrine tumor (panNET) and breast cancer models have demonstrated that nuclear-localized BCL-XL, rather than mitochondrial BCL-XL, drives epithelial-mesenchymal transition (EMT), migration, invasion, and stemness . Mechanistically, nuclear BCL-XL exerts these effects through epigenetic modification of the TGFβ promoter, thereby increasing TGFβ signaling . This was conclusively demonstrated using apoptosis-defective BCL-XL mutants and engineered BCL-XL targeted specifically to the nucleus, which retained pro-metastatic functions despite lacking anti-apoptotic activity. Further evidence comes from experiments with Bax/Bak double knockout mouse embryonic fibroblasts (MEFs), where BCL-XL overexpression significantly increased lung metastases formation (16.3±9.4 tumor foci per mouse compared to 2.3±1.2 in control, p=0.0046) despite the absence of Bax/Bak-mediated apoptosis . These findings highlight the importance of considering subcellular localization when studying BCL-XL functions in cancer progression.

What experimental models are most appropriate for studying BCL-XL in cancer research?

When selecting experimental models to study BCL-XL in cancer research, researchers should consider several complementary approaches. Cell line-based systems provide controlled conditions for mechanistic studies and allow genetic manipulation of BCL-XL expression. The N134 mouse panNET cell line and human colorectal cancer lines have been successfully used to demonstrate BCL-XL's role in invasion and migration . For more complex studies, three-dimensional organoid cultures better recapitulate tumor architecture and microenvironment interactions. In vivo models include:

  • Genetically engineered mouse models like RIP-Tag; RIP-tva for spontaneous multistep tumorigenesis, which revealed BCL-XL's role in invasion and metastasis of panNET

  • Xenograft models for experimental metastasis assays, as demonstrated with Bax/Bak double knockout MEFs overexpressing BCL-XL

  • Patient-derived xenografts that maintain tumor heterogeneity and microenvironment

For translational relevance, ex vivo tissue culture systems using freshly resected human tumor specimens, as employed in colorectal cancer studies, provide a bridge between preclinical models and clinical applications . Importantly, researchers should validate findings across multiple models, as BCL-XL's functions may be context-dependent across different cancer types.

How can researchers differentiate between BCL-XL's apoptotic and non-apoptotic functions in cancer cells?

To differentiate between BCL-XL's apoptotic and non-apoptotic functions, researchers should employ a multi-faceted experimental approach. One effective strategy is using subcellular targeting, where BCL-XL is engineered with specific localization signals directing it to the mitochondria (for apoptotic functions) or nucleus (for non-apoptotic functions like metastasis promotion) . Another approach involves structure-function studies using apoptosis-defective BCL-XL mutants that cannot bind pro-apoptotic BCL-2 family members but retain other functions .

Genetic models are particularly valuable, such as using Bax/Bak double knockout cells where the canonical apoptotic pathway is disabled, allowing isolated study of BCL-XL's non-apoptotic functions. This approach revealed that BCL-XL promotes lung metastasis formation even in apoptosis-deficient cells . Molecular pathway analysis through techniques like RNA-sequencing, ChIP-seq, and proteomics can identify BCL-XL's interaction partners and downstream effectors specific to each function. When assessing phenotypic outcomes, researchers should simultaneously measure multiple parameters: apoptosis markers (caspase activation, PARP cleavage), EMT markers (E-cadherin, vimentin), migration/invasion capability, and in vivo metastasis formation to comprehensively characterize BCL-XL's multifunctional impact on cancer progression.

What are the most effective approaches for targeting BCL-XL in cancer therapy?

Several strategies have been developed for targeting BCL-XL in cancer therapy, each with distinct advantages and considerations. Small molecule BH3 mimetics that competitively bind to the hydrophobic groove of BCL-XL, preventing its interaction with pro-apoptotic proteins, represent the most clinically advanced approach. These include navitoclax (ABT-263) which targets both BCL-2 and BCL-XL, and more selective BCL-XL inhibitors like A-1331852 and WEHI-539 . An innovative approach involves designing dual-binding proteins that simultaneously target BCL-XL and MCL-1, as demonstrated by computational protein design techniques that produced proteins with picomolar binding affinities (820 pM for BCL-XL and 196 pM for MCL-1) . This approach addresses the common problem of resistance to single-target inhibition.

Combining BCL-XL inhibition with standard chemotherapeutic agents has shown enhanced efficacy in preclinical colorectal cancer models, both in vitro and in ex vivo derived CRC tissue cultures . For cancers where nuclear BCL-XL drives metastasis, strategies targeting its non-canonical functions through disruption of epigenetic modifications or TGFβ signaling may prove beneficial . When implementing these approaches, researchers should carefully monitor dose-limiting thrombocytopenia, a common side effect of BCL-XL inhibition, and consider tumor-specific delivery systems to improve therapeutic index.

How can researchers address the challenge of thrombocytopenia when targeting BCL-XL?

Thrombocytopenia is a major dose-limiting toxicity when targeting BCL-XL therapeutically, as platelets depend on BCL-XL for survival. Researchers can address this challenge through several methodological approaches. Structural biology and medicinal chemistry can be employed to design BCL-XL inhibitors with modified pharmacokinetic properties that reduce platelet exposure while maintaining tumor penetration. Tumor-targeted delivery systems such as antibody-drug conjugates or nanoparticle formulations can direct BCL-XL inhibitors specifically to cancer cells while sparing platelets .

Intermittent dosing schedules with careful monitoring of platelet counts allow for platelet recovery between treatment cycles. Combination strategies using lower doses of BCL-XL inhibitors together with agents targeting complementary pathways can maintain efficacy while reducing thrombocytopenia risk. For experimental purposes, researchers should incorporate comprehensive hematological profiling in preclinical studies, including complete blood counts with particular attention to platelet numbers and function. Additionally, ex vivo studies using human platelets exposed to BCL-XL inhibitors can help predict thrombocytopenia risk before advancing to in vivo testing. These methodological considerations are essential for developing BCL-XL-targeting therapies with improved safety profiles.

What techniques are available for screening and validating BCL-XL inhibitors?

Researchers have multiple techniques available for screening and validating BCL-XL inhibitors across the drug development pipeline. For initial high-throughput screening, fluorescence polarization (FP) or time-resolved fluorescence resonance energy transfer (TR-FRET) assays measure displacement of fluorescently labeled BH3 peptides from recombinant BCL-XL protein. Surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC) provide detailed binding kinetics and thermodynamics, with demonstrated utility in characterizing picomolar-affinity interactions between designed proteins and BCL-XL .

Cellular validation includes mitochondrial outer membrane permeabilization (MOMP) assays, cytochrome c release assays, and measurement of caspase activation. The BH3 profiling technique can determine cellular dependence on specific anti-apoptotic proteins including BCL-XL. Cell viability assays should be performed in BCL-XL-dependent versus independent cell lines, with genetic knockdown/knockout controls to confirm specificity. For structural validation, X-ray crystallography has been successfully employed to confirm binding modes of designed inhibitors with BCL-XL, as demonstrated in crystallization studies using complexes at concentrations of approximately 23.5 mg/ml . In vivo validation requires monitoring tumor regression, survival benefit, and platelet counts in appropriate animal models. These complementary approaches ensure thorough characterization of BCL-XL inhibitors before clinical translation.

How does the subcellular localization of BCL-XL affect its function in different disease contexts?

At the endoplasmic reticulum, BCL-XL regulates calcium homeostasis by promoting Ca²⁺ transport to mitochondria, impacting cellular metabolism and ATP production . In neurons, BCL-XL localizes to synapses where it enhances synaptic plasticity and regulates neurotransmission, functions distinct from its neuroprotective role at mitochondria . To study these localization-dependent functions, researchers should employ subcellular fractionation followed by immunoblotting, immunofluorescence microscopy with colocalization analysis, and proximity ligation assays to detect interactions with compartment-specific partners. Manipulation of localization signals through site-directed mutagenesis or fusion with organelle-specific targeting sequences provides mechanistic insights into compartment-specific functions in disease pathogenesis.

What is the relationship between BCL-XL and autophagy in cancer cells?

The relationship between BCL-XL and autophagy in cancer cells is complex and context-dependent, involving both direct molecular interactions and indirect pathway crosstalk. BCL-XL directly interacts with Beclin 1, a key regulator of autophagy, through its BH3 domain . This interaction inhibits Beclin 1's ability to initiate autophagosome formation, thereby suppressing autophagy under normal conditions. During cellular stress, this inhibition can be relieved through competitive binding of pro-apoptotic BH3-only proteins or post-translational modifications of either protein, allowing coordinated regulation of both apoptosis and autophagy.

In cancer cells, this relationship becomes particularly significant as both processes influence treatment response. Inhibition of BCL-XL can induce not only apoptosis but also autophagic flux, which may serve as either a survival or death mechanism depending on cancer type and treatment context. To investigate this relationship, researchers should employ multiple methodological approaches, including co-immunoprecipitation to detect BCL-XL-Beclin 1 binding, autophagy flux assays using LC3-II and p62 markers, and genetic manipulation of both pathways simultaneously. Live-cell imaging with fluorescently tagged BCL-XL and autophagy markers provides dynamic insights into their spatial and temporal relationship during treatment response. Understanding this interplay is crucial for designing therapeutic strategies that effectively target BCL-XL-dependent cancers while navigating potential autophagy-mediated resistance mechanisms.

How can computational approaches advance BCL-XL research and drug development?

Computational approaches have become increasingly valuable in BCL-XL research, offering powerful tools for understanding protein interactions and designing targeted therapeutics. Structure-based computational design has successfully produced proteins that simultaneously bind BCL-XL and MCL-1 with picomolar affinities (820 pM and 196 pM, respectively) . This was achieved using sophisticated algorithms implemented in the Rosetta software suite, including the BundleGridSampler mover to generate structural elements and FastDesign mover for sequence optimization . These computationally designed proteins demonstrated potent apoptosis-inducing activity in cancer cells dependent on both BCL-XL and MCL-1.

For gene expression analysis, computational approaches have revealed BCL-XL as the only antiapoptotic BCL-2 protein overactivated in colorectal cancer through RNA sequencing analysis of >1500 patients, followed by prediction of protein activity . In diagnostic applications, bioinformatic analysis demonstrated BCL-XL's excellent discrimination capacity (AUC: 0.83 [95% CI: 0.76, 0.90]) for pancreatic ductal adenocarcinoma versus normal tissue .

Researchers can leverage molecular dynamics simulations to study conformational changes in BCL-XL upon binding different partners, and machine learning approaches to predict drug sensitivity based on BCL-XL expression patterns. Network analysis algorithms can identify synthetically lethal partners of BCL-XL for combination therapy strategies. These computational methods, when integrated with experimental validation, substantially accelerate BCL-XL research and therapeutic development.

How do different viruses modulate BCL-XL expression and function?

Viruses have evolved diverse mechanisms to modulate BCL-XL expression and function to facilitate their replication and persistence. Multiple RNA and DNA viruses interact with BCL-XL, including hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), influenza A virus (IAV), Epstein-Barr virus (EBV), human T-lymphotropic virus type-1 (HTLV-1), Maraba virus (MRBV), Schmallenberg virus (SBV), and coronavirus (CoV) . These viruses can either upregulate or downregulate BCL-XL depending on their replication strategy and lifecycle stage.

Some viruses, particularly those establishing chronic infections like HBV, HCV, and EBV, often upregulate BCL-XL to prevent premature death of infected cells, ensuring viral persistence and production. This upregulation can occur through direct viral protein interactions with cellular transcription factors or through activation of signaling pathways such as NF-κB or STAT3 . In contrast, certain lytic viruses may downregulate BCL-XL during late infection stages to facilitate cell lysis and viral release. The modulation of BCL-XL by viruses has significant implications for viral pathogenesis, including tissue damage, oncogenesis in the case of oncogenic viruses, and the establishment of persistent infections. Researchers studying these interactions should employ time-course experiments to capture the dynamic regulation of BCL-XL throughout the viral lifecycle and utilize virus-specific systems to account for tropism differences.

What methodological approaches can researchers use to study BCL-XL's role in viral pathogenesis?

To comprehensively study BCL-XL's role in viral pathogenesis, researchers should employ multiple complementary methodological approaches. For in vitro studies, time-course experiments tracking BCL-XL expression levels during viral infection using qRT-PCR, Western blotting, and immunofluorescence microscopy reveal dynamic regulation patterns. Genetic manipulation through CRISPR-Cas9 knockout or siRNA knockdown of BCL-XL in permissive cell lines, followed by viral infection, helps establish causal relationships between BCL-XL levels and viral replication efficiency or cytopathic effects .

Pharmacological approaches using specific BCL-XL inhibitors at different stages of viral infection can identify critical windows where BCL-XL function impacts viral lifecycle. For studying protein-protein interactions, co-immunoprecipitation and proximity ligation assays can detect direct interactions between viral proteins and BCL-XL or other apoptotic machinery components. In vivo models, including BCL-XL conditional knockout mice infected with relevant viruses, provide systemic context for understanding BCL-XL's role in viral pathogenesis, immune responses, and tissue damage.

Translational studies should examine BCL-XL expression in infected human tissues through immunohistochemistry and correlate with viral load, disease severity, and clinical outcomes. High-throughput screening of BCL-XL modulators may identify compounds with antiviral activity through disruption of virus-dependent BCL-XL regulation. These methodological approaches collectively provide a comprehensive understanding of how BCL-XL influences viral pathogenesis and identify potential therapeutic targets.

How can targeting BCL-XL be incorporated into antiviral therapeutic strategies?

Targeting BCL-XL represents a promising adjunctive approach for antiviral therapies, particularly for viruses that modulate apoptotic pathways to facilitate replication or persistence. For viruses that upregulate BCL-XL to prevent infected cell death, selective BCL-XL inhibitors (such as A-1331852 or WEHI-539) could promote elimination of infected cells through restoration of apoptotic sensitivity . This approach may be particularly valuable against persistent viral infections like hepatitis B virus (HBV), hepatitis C virus (HCV), and Epstein-Barr virus (EBV), where infected cell survival contributes to chronicity and associated pathologies.

Implementation of this strategy requires careful consideration of several methodological aspects. Timing of BCL-XL inhibition is critical—early administration may limit viral spread, while delayed administration might optimize clearance of infected cells. Combination approaches pairing BCL-XL inhibitors with direct-acting antivirals could synergistically reduce viral load while eliminating infected cell reservoirs. To mitigate potential toxicities, especially thrombocytopenia, researchers should consider localized delivery systems targeting infected tissues or pulsed dosing schedules.

When designing studies to evaluate this approach, researchers should incorporate comprehensive endpoints measuring not only viral load reduction but also infected cell clearance, tissue damage assessment, and immune response characterization. Appropriate in vitro and animal models that recapitulate key aspects of human viral infections are essential for preclinical evaluation before translation to clinical studies. This integrated strategy leveraging BCL-XL inhibition could complement conventional antiviral approaches by addressing the cellular reservoirs that often contribute to treatment failure.

What emerging technologies will advance our understanding of BCL-XL biology?

Several cutting-edge technologies are poised to transform BCL-XL research in the coming years. Single-cell multi-omics approaches combining transcriptomics, proteomics, and metabolomics at the individual cell level will reveal previously unappreciated heterogeneity in BCL-XL expression and function across cell populations. This will be particularly valuable for understanding BCL-XL's role in tumor heterogeneity and treatment resistance . CRISPR-based functional genomics screens, including CRISPR activation and interference (CRISPRa/CRISPRi), will enable systematic identification of genes that synthetically interact with BCL-XL, revealing novel therapeutic vulnerabilities and resistance mechanisms.

Advanced structural biology techniques such as cryo-electron microscopy (cryo-EM) will provide dynamic visualizations of BCL-XL interactions with binding partners in near-native conditions, offering insights beyond static crystal structures . Protein-protein interaction mapping through BioID or APEX proximity labeling will reveal the complete BCL-XL interactome across different subcellular compartments, helping elucidate its diverse functions beyond apoptosis regulation .

In vivo imaging technologies using genetically encoded reporters will allow real-time tracking of BCL-XL activity in living organisms during disease progression and treatment response. Advanced computational approaches, including deep learning algorithms, will integrate multi-dimensional data to predict BCL-XL-dependent phenotypes and treatment responses. These emerging technologies will collectively drive forward our understanding of BCL-XL biology and accelerate the development of more effective therapeutic strategies.

How can researchers address the challenge of contextual dependencies in BCL-XL function?

Addressing the contextual dependencies in BCL-XL function requires systematic methodological approaches that capture the complexity of its behavior across different cellular environments. Researchers should implement tissue-specific and inducible genetic models that allow precise spatial and temporal control of BCL-XL expression and localization. This includes conditional knockout/knockin mice and cell-type-specific promoters for in vitro studies that can reveal how the same BCL-XL-targeting intervention produces different outcomes across tissue contexts .

Multi-parametric phenotypic profiling using high-content imaging and flow cytometry enables simultaneous measurement of multiple cellular responses to BCL-XL modulation, capturing contextual differences in a high-dimensional space. Systems biology approaches integrating transcriptomic, proteomic, and metabolomic data can identify context-specific networks and dependencies associated with BCL-XL function. These should be coupled with computational modeling to predict how cellular context influences BCL-XL behavior and treatment response.

In the experimental design phase, researchers should systematically vary key contextual parameters (oxygen tension, nutrient availability, extracellular matrix composition, cell-cell interactions) to build comprehensive response maps. Patient-derived models including organoids and xenografts preserve the native cellular context and heterogeneity, allowing more physiologically relevant study of BCL-XL dependencies . Through these methodological approaches, researchers can develop a more nuanced understanding of BCL-XL's context-dependent functions and design more precisely targeted therapeutic strategies.

What are the key considerations for translating BCL-XL research from bench to bedside?

Translating BCL-XL research from bench to bedside requires careful consideration of several critical factors to ensure safety and efficacy in clinical applications. Patient stratification is paramount—researchers must develop robust biomarkers to identify patients whose tumors are truly dependent on BCL-XL. This includes not only expression levels but functional dependence, potentially assessed through BH3 profiling or gene expression signatures . Given BCL-XL's role in normal tissues, particularly platelets, therapeutic window optimization is essential. This involves designing dosing strategies, drug delivery systems, or combination approaches that maximize tumor impact while minimizing toxicity to normal tissues.

Resistance mechanism anticipation is crucial, as cancer cells can adapt to BCL-XL inhibition through various pathways. Preclinical studies should proactively investigate potential resistance mechanisms and develop strategies to prevent or overcome them, such as rational combinations targeting multiple BCL-2 family proteins simultaneously . For clinical trial design, researchers should incorporate pharmacodynamic endpoints beyond tumor shrinkage, including on-target effects (BAX/BAK activation, cytochrome c release) and functional imaging to assess early response.

Regulatory considerations include careful toxicity profiling, with special attention to hematological parameters, and designing first-in-human studies with appropriate safety monitoring. Throughout the translation process, continuous refinement of understanding BCL-XL biology should inform clinical development, with bidirectional flow of information between laboratory and clinic. This integrated approach will maximize the potential for successful translation of BCL-XL-targeted therapies into clinical benefit.

Product Science Overview

Introduction

B-Cell Lymphoma Extra Large (BCL-xL) is a member of the BCL-2 protein family, which plays a crucial role in regulating cell death and survival. BCL-xL is encoded by the BCL2L1 gene and is known for its anti-apoptotic properties, making it a significant target in cancer research and therapy. The recombinant form of BCL-xL, produced through genetic engineering, allows for detailed studies and potential therapeutic applications.

BCL-2 Protein Family

The BCL-2 protein family consists of several proteins that regulate apoptosis, a form of programmed cell death essential for maintaining cellular homeostasis. The family is divided into three subgroups based on their function and structure:

  1. Anti-apoptotic proteins: These include BCL-2, BCL-xL, and MCL-1, which prevent apoptosis by inhibiting pro-apoptotic proteins.
  2. Pro-apoptotic proteins: These include BAX and BAK, which promote apoptosis by forming pores in the mitochondrial membrane.
  3. BH3-only proteins: These include BID, BIM, and PUMA, which regulate the activity of both anti-apoptotic and pro-apoptotic proteins.
Structure and Function of BCL-xL

BCL-xL is a mitochondrial protein that inhibits apoptosis by preventing the release of cytochrome c from the mitochondria, a critical step in the apoptotic pathway. It achieves this by binding to and neutralizing pro-apoptotic proteins such as BAX and BAK. BCL-xL has also been implicated in other cellular processes, including autophagy and senescence, where it can have dual roles depending on the cellular context .

Role in Cancer

BCL-xL is overexpressed in various cancers, including B-cell lymphomas, where it contributes to tumor cell survival and resistance to chemotherapy. Its anti-apoptotic function allows cancer cells to evade programmed cell death, leading to uncontrolled cell proliferation. Targeting BCL-xL with specific inhibitors is a promising therapeutic strategy in cancer treatment .

Recombinant BCL-xL

The recombinant form of BCL-xL is produced using recombinant DNA technology, where the BCL2L1 gene is inserted into an expression vector and introduced into host cells, such as bacteria or yeast. These host cells then produce the BCL-xL protein, which can be purified and used for research or therapeutic purposes. Recombinant BCL-xL allows for detailed structural and functional studies, aiding in the development of targeted therapies.

Therapeutic Applications

Given its role in cancer cell survival, BCL-xL is a potential target for cancer therapy. Several small-molecule inhibitors of BCL-xL have been developed, aiming to restore the apoptotic pathway in cancer cells. These inhibitors can sensitize cancer cells to chemotherapy and improve treatment outcomes. Additionally, understanding the dual roles of BCL-xL in autophagy and senescence may provide insights into novel therapeutic approaches .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.