Annexin A5 (ANXA5), a calcium-dependent phospholipid-binding protein, plays diverse roles in cellular processes such as membrane repair, apoptosis regulation, and immune modulation. In murine models, ANXA5 has been extensively studied to elucidate its physiological and pathological functions, particularly in cardiovascular, auditory, and immune systems. This article synthesizes key findings from peer-reviewed studies focusing on ANXA5 Mouse, encompassing its abundance, functional roles, and therapeutic potential.
ANXA5 is highly abundant in specific mouse tissues, particularly in sensory and vascular systems.
ANXA5 is the most abundant membrane-associated protein in mouse vestibular hair bundles, with ~15,000 copies per stereocilium in young adult mice (P23). This concentration exceeds other annexin isoforms, which collectively account for only 1,000 molecules per stereocilium . Despite its abundance, Anxa5 knockout (−/−) mice exhibit normal hearing and balance, suggesting functional redundancy or compensatory mechanisms .
Developmental Stage | ANXA5 Copies/Stereocilium | Relative Abundance |
---|---|---|
P5 (Developing) | ~3,500 | Most abundant annexin |
P23 (Young Adult) | ~15,000 | Dominates over other annexins |
Cochlea: Concentrated at stereocilia tips in inner and outer hair cells by P28 .
Utricle: Detected in hair bundles and somas of hair cells and supporting cells .
ANXA5 administration has shown therapeutic potential in myocardial infarction (MI) and atherosclerosis.
In a mouse model of MI-reperfusion injury, intraperitoneal AnxA5 treatment reduced infarct size by 27% and improved cardiac function (e.g., 29% increase in LV ejection fraction) . Key outcomes:
Parameter | Vehicle Treatment | AnxA5 Treatment |
---|---|---|
Infarct Size (MRI) | Baseline | 27% reduction |
Macrophages (Infarct Area) | 306.4 ± 63.1/mm² | 134.5 ± 21.3/mm² |
LV Fibrosis | Baseline | 42% reduction |
Mechanisms include reduced inflammatory cell infiltration and enhanced plaque stability .
In ApoE−/− mice, early AnxA5 administration (1 mg/kg, thrice weekly) reduced plaque size in the aortic root (36% reduction) and arch (55% reduction) .
Atherosclerosis Model | Plaque Parameter | Vehicle | AnxA5 |
---|---|---|---|
Aortic Root (6 weeks) | Size (μm²) | 130,027 ± 16,988 | 83,766 ± 9,408 |
Aortic Arch (6 weeks) | Size (μm²) | 221,811 ± 39,118 | 99,770 ± 9,922 |
ANXA5 acts as an immune checkpoint inhibitor by binding phosphatidylserine (PS) on apoptotic cells, preventing immunosuppressive interactions.
In chemotherapy-induced apoptosis, AnxA5 administration enhances antitumor immunity by blocking PS-mediated immunosuppression. Fusion of AnxA5 with tumor antigens improves vaccine efficacy in murine cancer models .
Therapeutic Approach | Outcome |
---|---|
AnxA5 + Chemotherapy | Reduced tumor growth, enhanced immunity |
AnxA5-Antigen Fusion | Targeted antigen delivery to tumors |
Despite high expression in hair bundles, Anxa5 −/− mice show no deficits in hearing or balance. This suggests ANXA5 may have non-critical roles in mechanotransduction or compensate via other annexins .
Recombinant mouse ANXA5 (produced in E. coli) is used in therapeutic studies. Key properties :
Property | Description |
---|---|
Molecular Weight | 38.1 kDa |
Purification | Chromatographic techniques |
Formulation | PBS, 10% glycerol, 1 mM DTT |
Stability | Store at -20°C for long-term use |
ANXA5 (Annexin A5) is a single-chain phospholipid-binding protein belonging to the annexin gene superfamily. In mice, it is encoded by the Anxa5 gene with accession number P48036 . The recombinant mouse ANXA5 protein has a calculated molecular mass of approximately 36 kDa, with its expressed region typically spanning from Ala2 to Asp319 . ANXA5 is characterized by its high-affinity binding to phosphatidylserine (PS) in a calcium-dependent manner, which is central to its biological functions . The protein is also known by several synonyms including Annexin V, Anchorin CII, Calphobindin I (CBP-I), Lipocortin V, and Vascular anticoagulant-alpha (VAC-alpha) .
Research indicates significant changes in ANXA5 abundance during mouse development, particularly in specialized tissues. In vestibular hair bundles of the inner ear, ANXA5 concentration increases substantially from developmental stages to adulthood. Quantitative mass spectrometry has shown that in postnatal days 4-6 (P5) mouse utricle hair bundles, there are approximately 3,500 ANXA5 molecules per stereocilium, whereas by P21-P25 ("P23"; young adult), this increases dramatically to approximately 15,000 molecules per stereocilium . This developmental increase suggests stage-specific functions that may relate to membrane stabilization or repair mechanisms as the sensory structures mature. Unlike many developmental changes, this increase is not compensated by other annexin family members, as their combined abundance remains at approximately 1,000 molecules per stereocilium regardless of developmental stage .
While the search results don't directly compare mouse and human ANXA5, research protocols reveal important insights. Studies often use human ANXA5 cDNA expressed in bacterial systems like Escherichia coli M15 for experimental applications in mouse models . This cross-species compatibility suggests significant structural and functional conservation between mouse and human ANXA5. Both function similarly in phosphatidylserine binding and show parallel anti-inflammatory properties when administered in mouse disease models . The effectiveness of human ANXA5 in mouse experimental systems indicates that the core functional domains and mechanisms are highly conserved between species, making mouse models valuable for translational research investigating ANXA5's therapeutic potential in human diseases.
Recombinant mouse ANXA5 can be produced through several expression systems, with mammalian and bacterial systems being the most common. For studies requiring post-translational modifications, HEK293 cell expression systems are often preferred. The protocol involves:
Transfecting HEK293 cells with an expression vector harboring the mouse ANXA5 gene
Growing cells in serum-free medium
Harvesting cell culture by centrifugation to remove cells
Collecting the supernatant containing secreted ANXA5
Adding mammalian cell protease inhibitor cocktail
Immediate aliquoting and storage at -80°C
For higher yield and less expensive production, bacterial expression systems can be used:
Transform Escherichia coli M15 (pREP4) with pQE30Xa containing mouse ANXA5 cDNA
Harvest bacteria and lyse by sonification
Remove cell debris by centrifugation
Isolate His-tagged ANXA5 from supernatant using nickel columns and an imidazole gradient
Verify protein homogeneity using MALDI-TOF/TOF
Confirm PS binding activity through ellipsometry
Ensure endotoxin levels are below one endotoxin unit per ml using spectrophotometry
Both methods produce functional ANXA5, though the choice depends on experimental requirements for post-translational modifications and application.
Quantitative mass spectrometry has proven particularly effective for precise measurement of ANXA5 expression levels in mouse tissues. The recommended methodology includes:
Tissue isolation (e.g., bundle isolation from mouse utricles using the twist-off technique)
Sample preparation for shotgun mass spectrometry
Analysis using an Orbitrap mass spectrometer
Quantification with MaxQuant software and the iBAQ algorithm
Measurement of peptide extracted ion chromatogram areas
Calculation of relative molar abundance in specific tissues or cellular compartments
This approach allows researchers to determine that ANXA5 accounts for approximately 2% of total protein in mouse vestibular hair bundles, representing about 15,000 copies per stereocilium . For relative quantification across multiple annexin family members, this method enables comprehensive profiling that reveals ANXA5 as the most abundant membrane-associated protein in certain structures, far exceeding other annexin isoforms .
While the search results don't provide a comprehensive protocol for generating Anxa5 knockout mice, they do reference the use of Anxa5^-/- mice in research, suggesting established methodologies exist . Based on standard practices and the referenced studies, an effective approach would include:
Generation phase:
Use gene targeting techniques to disrupt the Anxa5 gene
Confirm targeting through Southern blotting and PCR genotyping
Establish breeding colonies with appropriate background strains
Validation phase:
Functional validation:
Test physiological functions related to known ANXA5 roles (hearing, balance, cardiovascular function)
Challenge mice with appropriate stressors (e.g., noise exposure for hearing studies)
The search results indicate that properly validated Anxa5^-/- mice show normal development, no mortality differences, and similar weight progression compared to wild-type mice, suggesting effective compensation mechanisms operate in the absence of this protein .
Research using ApoE^-/- mouse models provides substantial evidence for ANXA5's protective role in atherosclerosis. Administration of ANXA5 during early-onset atherosclerosis significantly reduces plaque formation and promotes plaque stability. Key experimental findings include:
Reduced atherosclerotic development in the aortic root (130,027 ± 16,988 vs. 83,766 ± 9,408 μm², p = 0.0242) and aortic arch (221,811 ± 39,118 vs. 99,770 ± 9,922 μm², p = 0.0077) compared to control animals
Decreased plaque formation consistently across all vessels in the aortic arch (excluding the carotid artery), suggesting a generalized mechanism rather than a flow-associated effect
Reduced macrophage infiltration and increased expression of plaque stability markers in animals receiving ANXA5 treatment
Diminished rate of apoptosis within plaques during early disease stages, though this effect appears to be overwhelmed in later stages of atherosclerosis
These findings suggest a dual protective mechanism: early-stage reduction in apoptotic rates and macrophage infiltration, combined with inhibition of monocyte adhesion to the endothelial layer, collectively reducing plaque growth and promoting stability .
Based on experimental protocols with demonstrated efficacy, the optimal dosing regimen for ANXA5 in mouse atherosclerosis models consists of:
Dose: 1 mg/kg body weight
Administration route: Intraperitoneal (i.p.) injection
Frequency: Three times weekly
Treatment initiation: After one week of Western-type diet (0.25% cholesterol)
Treatment duration: Six weeks has shown significant effects on plaque development
This regimen was established based on blood clearance studies determining ANXA5's half-life following i.p. injection to be 5.64 ± 2.33 hours . The dosage and administration schedule are designed to maintain therapeutic levels while accounting for clearance rates. This protocol has proven effective in reducing plaque formation in the ApoE^-/- mouse model without causing adverse effects on weight development, mortality, cholesterol levels, or circulating leukocyte subsets .
Contrary to what might be expected given ANXA5's abundance in inner ear structures, research has shown that ANXA5 does not play a significant role in protecting against noise-induced hearing damage in mouse models. Despite ANXA5 being the most abundant membrane-associated protein in vestibular hair bundles (~15,000 copies per stereocilium), mature Anxa5^-/- mice show:
Normal hearing and balance function under standard conditions
Identical responses to noise exposure compared to wild-type mice
No differences in temporary or permanent changes in hearing sensitivity following acoustic trauma
These findings are particularly surprising given annexins' proposed role in calcium-dependent repair of membrane lesions, which could theoretically contribute to recovery mechanisms after noise damage. The research suggests that despite its unusual abundance, ANXA5 does not participate in the hair bundle's mechanotransduction function or in protective responses to acoustic trauma, at least until after two months of age in the cochlea and six months in the vestibular system .
The high abundance yet limited functional significance of ANXA5 in certain tissues presents an intriguing research paradox. To address this scientifically, researchers should consider the following methodological approaches:
Age-dependent functional analyses:
Stress-specific challenges:
Test beyond acoustic trauma by examining responses to other stressors (oxidative stress, mechanical trauma, ototoxic compounds)
Implement multiple stress paradigms with varying intensities and durations
Subtler phenotypic analyses:
Develop more sensitive assays for vestibular and auditory function beyond standard tests
Implement single-cell electrophysiology to detect subtle changes in mechanotransduction
Use high-resolution imaging to identify subcellular structural alterations
Redundancy and compensation mechanisms:
Systematically inhibit multiple annexin family members simultaneously
Create conditional knockouts with temporal control to bypass developmental compensation
Perform comprehensive transcriptomic and proteomic analyses to identify upregulated compensatory pathways in ANXA5's absence
This scientific paradox reinforces the critical concept that protein abundance does not necessarily correlate with functional significance within specific cellular structures or compartments .
Investigating ANXA5's calcium-dependent membrane binding in live mouse tissues requires sophisticated methodological approaches that balance physiological relevance with technical feasibility:
Calcium concentration control:
Implement precise calcium buffering systems calibrated to physiological ranges
Use calcium ionophores to experimentally manipulate calcium levels in a controlled manner
Employ genetically-encoded calcium indicators for spatiotemporal analysis of calcium fluctuations
Membrane dynamics visualization:
Use fluorescently-tagged ANXA5 constructs (ensuring tag doesn't interfere with binding)
Implement FRAP (Fluorescence Recovery After Photobleaching) to measure binding kinetics
Apply super-resolution microscopy techniques to visualize nanoscale membrane interactions
Phosphatidylserine accessibility monitoring:
Use complementary PS-binding probes with different binding properties
Implement systems to induce controlled PS exposure (e.g., apoptosis inducers, membrane scrape techniques)
Consider dual-labeling approaches to simultaneously track PS exposure and ANXA5 binding
Tissue-specific considerations:
For hair cells, develop methods to maintain viability of isolated stereocilia or hair bundles
For vascular tissues, consider ex vivo flow systems that maintain physiological pressure and flow
Develop clearing techniques compatible with calcium-dependent binding studies for whole-tissue imaging
Quantification approaches:
Implement ratiometric imaging for quantitative binding analysis
Develop computational models to estimate binding parameters from imaging data
Use advanced image analysis algorithms to detect subtle changes in membrane association patterns
These methodological considerations address the technical challenges of studying dynamic calcium-dependent processes while maintaining physiological relevance.
To ensure specificity in ANXA5 binding studies using mouse models, researchers should implement a comprehensive set of controls addressing multiple aspects of experimental design:
Genetic controls:
Include Anxa5^-/- mice as negative controls
Use wild-type littermates as positive controls
Consider heterozygous mice to evaluate dose-dependent effects
Protein-specific controls:
Calcium-dependency controls:
Perform parallel experiments with calcium chelators (EGTA)
Include calcium titration series to establish dose-response relationships
Implement calcium-free conditions as negative controls
Phospholipid specificity controls:
Use liposomes with defined phospholipid compositions
Include membranes lacking phosphatidylserine as negative controls
Implement competition with other PS-binding proteins (e.g., lactadherin)
Technical controls:
For labeled ANXA5, include the fluorophore/tag alone to control for non-specific interactions
Perform pre-adsorption controls to verify antibody specificity
Include isotype controls for immunological detection methods
Sample preparation controls:
Process tissues from different mouse strains identically
Implement vehicle-only controls for any treatments
Include time-matched controls to account for potential temporal variations
These controls collectively ensure that observed effects are specifically attributable to ANXA5's physiological binding properties rather than experimental artifacts or non-specific interactions.
When faced with contradictory findings between in vitro and in vivo ANXA5 studies in mouse models, researchers should employ a systematic analytical approach:
Contextual analysis:
Examine physiological relevance of in vitro conditions (calcium concentrations, PS accessibility)
Consider developmental timing differences between systems
Evaluate the complexity of the microenvironment present in vivo but absent in vitro
Methodological reconciliation:
Compare protein concentrations between systems (local concentrations in vivo may differ significantly)
Assess differences in post-translational modifications between systems
Evaluate temporal aspects (acute vs. chronic effects)
Integrated analysis framework:
Consider in vitro findings as mechanistic insights rather than functional predictions
View in vivo results as net effects that may mask opposing mechanisms
Develop intermediate complexity models (ex vivo systems, organoids) to bridge the gap
Specific examples from ANXA5 research:
In vitro studies show ANXA5 binding to PS-expressing cells suppresses pro-inflammatory actions
Flow chamber experiments demonstrate AnxA5 inhibits monocyte adhesion by 20%
Yet Anxa5^-/- mice show normal development and function in certain systems
This suggests compensatory mechanisms operate in vivo that are absent in vitro, or that the threshold for functional significance differs between systems.
Translational implications:
Consider pharmacological effects (administered ANXA5) versus genetic models (knockout)
Evaluate dose-responses that may explain different outcomes
Assess bioavailability and tissue distribution factors present in vivo but not in vitro
This analytical framework helps researchers synthesize seemingly contradictory findings into a more comprehensive understanding of ANXA5 biology.
Quantifying ANXA5 expression across different mouse tissues presents several technical challenges that can be addressed through methodological refinements:
Challenge | Technical Solution | Implementation Notes |
---|---|---|
Tissue-specific extraction efficiency | Develop tissue-specific extraction protocols | Optimize lysis buffers for each tissue type; validate recovery with spiked-in standards |
Variable protein-to-lipid ratios | Normalize to multiple reference markers | Use both membrane and cytosolic markers; implement absolute quantification methods |
Post-translational modifications | Use multiple peptides across protein sequence | Select peptides from conserved regions avoiding known modification sites |
Cross-reactivity with other annexins | Employ isoform-specific detection methods | Use mass spectrometry with unique peptide identification; validate antibody specificity |
Subcellular compartmentalization | Implement fractionation before quantification | Develop protocols for isolation of relevant compartments (e.g., membrane vs. cytosol) |
Low abundance in certain tissues | Employ enrichment strategies | Use immunoprecipitation or targeted mass spectrometry approaches |
Dynamic temporal expression | Standardize tissue collection timing | Control for circadian effects; use consistent developmental timepoints |
For optimal results, researchers should implement: |
Mass spectrometry-based quantification using the iBAQ algorithm for absolute quantification
Western blotting with carefully validated antibodies for relative expression
Parallel mRNA quantification to assess transcriptional versus post-transcriptional regulation
Preparation of reference standards from recombinant mouse ANXA5 for calibration curves
Spatial mapping using immunohistochemistry to complement quantitative data
These approaches collectively address the technical challenges in obtaining reliable quantitative data across diverse tissue contexts.
When analyzing ANXA5 effects in atherosclerosis progression studies using mouse models, researchers should implement statistical approaches that address the specific characteristics of these experiments:
Power analysis considerations:
Primary outcome measures:
For plaque size: unpaired t-tests or Mann-Whitney tests depending on normality
For multiple vessel comparisons: ANOVA with appropriate post-hoc tests, with correction for multiple comparisons
For categorical data: Chi-square or Fisher's exact test
Longitudinal data analysis:
Mixed-effects models to account for repeated measures
Analysis of rate of progression rather than endpoint measurements
Time-to-event analysis for development of advanced plaques
Addressing covariates:
ANCOVA to control for variables like weight, cholesterol levels, and baseline measurements
Multivariate regression to identify predictors of treatment response
Stratified analysis when appropriate (e.g., by sex or baseline plaque burden)
Dose-response relationships:
Implement dose-response modeling (linear, sigmoidal) when multiple doses are tested
Calculate EC50 values for therapeutic applications
Test for linearity of response
Reporting requirements:
Always include exact p-values rather than thresholds
Report confidence intervals for primary outcome measures
Present individual data points alongside means and standard deviations
Clearly state the statistical tests used for each analysis
These statistical approaches provide robust analysis of ANXA5 effects while accounting for the biological variability inherent in atherosclerosis studies.
Given the lack of phenotype in Anxa5^-/- mice and evidence of limited compensation by other annexins, several promising research directions emerge:
Systematic combinatorial knockout studies:
Generate double or triple knockouts of annexin family members
Prioritize combinations based on co-expression patterns
Implement conditional knockout systems to bypass embryonic lethality
Stress-response profiling:
Subject Anxa5^-/- mice to various stressors (oxidative, mechanical, inflammatory)
Perform comprehensive transcriptomic and proteomic analyses under stress conditions
Identify stress-specific compensatory mechanisms
Functional domain analysis:
Create knock-in mice expressing chimeric annexins (domain swapping)
Test whether specific domains rather than whole proteins provide functional redundancy
Identify critical residues for shared functions through point mutations
Temporal dynamics investigations:
Implement time-resolved proteomics following annexin knockout
Study developmental switches in annexin expression and function
Use inducible knockout systems to distinguish developmental from acute compensation
Cell-type specific analyses:
Perform single-cell transcriptomics to identify cell populations with unique annexin expression patterns
Create cell-type specific knockouts to bypass systemic compensation
Investigate annexin redundancy in specialized cell types (e.g., hair cells, macrophages)
Evolution-guided approaches:
Conduct comparative studies across species with varying annexin repertoires
Identify evolutionary patterns suggesting functional redundancy
Test whether annexin functions are conserved across distant species
These research directions would significantly advance our understanding of functional redundancy within the annexin family, potentially revealing context-specific roles that aren't apparent in standard knockout models.
Advanced imaging techniques offer transformative potential for understanding ANXA5 dynamics in living mouse tissues:
Intravital microscopy applications:
Implement cranial windows for real-time imaging of ANXA5 in cerebrovascular disease models
Develop abdominal imaging windows for visualizing ANXA5 in atherosclerosis progression
Use genetically encoded calcium indicators alongside fluorescently tagged ANXA5 to correlate calcium dynamics with binding events
Super-resolution approaches:
Apply STED microscopy to resolve ANXA5 clustering at nanoscale resolution
Implement PALM/STORM imaging to track single-molecule dynamics at the membrane
Use expansion microscopy to map ANXA5 distribution within complex tissue architectures
Functional correlation techniques:
Combine electrophysiology with fluorescence imaging in hair cells
Implement tension sensors to correlate mechanical forces with ANXA5 recruitment
Use optogenetic calcium manipulation to trigger and observe ANXA5 translocation events
Multi-parameter imaging strategies:
Implement multiplexed imaging to simultaneously track multiple annexin family members
Combine phosphatidylserine probes with ANXA5 imaging to correlate lipid dynamics
Integrate membrane tension sensors with ANXA5 imaging to test mechanosensitive binding
Temporal resolution enhancements:
Apply fluorescence lifetime imaging to distinguish bound from unbound ANXA5
Use high-speed imaging to capture rapid membrane repair events
Implement light-sheet microscopy for extended time-lapse imaging with minimal phototoxicity
Whole-animal imaging approaches:
Develop near-infrared ANXA5 probes for non-invasive imaging of inflammation
Apply PET imaging with radiolabeled ANXA5 to track distribution in disease models
Implement photoacoustic imaging for deeper tissue visualization of ANXA5 dynamics
These advanced imaging approaches would provide unprecedented insights into the dynamic behavior of ANXA5 in living tissues, potentially revealing functional roles that have been missed by endpoint analyses.
Based on current understanding of ANXA5's properties, several emerging therapeutic applications in mouse models of human disease show promise:
Atherosclerosis and cardiovascular applications:
Inflammatory disease modulation:
Investigate ANXA5 for reducing inflammation in autoimmune disease models
Test PS-binding properties for neutralizing inflammatory extracellular vesicles
Develop combination approaches targeting multiple danger-associated molecular patterns (DAMPs)
Membrane repair applications:
Engineer ANXA5 variants with enhanced membrane binding properties
Test preventive administration before exposure to membrane-damaging agents
Investigate tissue-specific delivery for organs vulnerable to membrane damage
Diagnostic imaging platforms:
Develop ANXA5-based molecular imaging agents for detecting apoptosis in vivo
Create multimodal probes combining ANXA5 with various imaging reporters
Test ANXA5 imaging for early detection of treatment response in cancer models
Extracellular vesicle engineering:
Use ANXA5 coating to reduce immunogenicity of therapeutic vesicles
Develop ANXA5-based purification systems for isolating specific vesicle populations
Create ANXA5 fusion proteins for targeting therapeutic vesicles to specific tissues
Biomaterial applications:
Incorporate ANXA5 into biomaterials to improve biocompatibility
Develop ANXA5-coated implants for reduced inflammatory response
Create smart materials with calcium-dependent ANXA5 presentation These therapeutic directions build upon ANXA5's unique properties while addressing limitations in current approaches, potentially leading to novel interventions for human diseases that have been validated in mouse models.
Annexin A5, also known as ANXA5 or annexin V, is a cellular protein belonging to the annexin family. This family comprises over 160 proteins characterized by their ability to bind to negatively charged phospholipid surfaces in a calcium-dependent manner . Annexin A5 is widely studied for its role in various biological processes, including apoptosis, blood coagulation, and cellular signaling.
Annexin A5 was first isolated from human placenta in the late 1970s . The protein has a molecular weight of approximately 35 kDa and consists of four annexin repeats, which are responsible for its calcium-dependent phospholipid-binding properties . The recombinant form of Annexin A5, derived from mouse, is commonly used in research to study its functions and applications.
Annexin A5 has significant potential in clinical applications, particularly in the fields of cardiology and oncology. Radiolabeled Annexin A5 can be used as a diagnostic tool to visualize cell death in conditions such as myocardial infarction, atherosclerosis, and cancer . Additionally, it may serve as a therapeutic agent for monitoring and targeting cell death-inducing therapies.
In the laboratory, Annexin A5 is commonly used in flow cytometry and fluorescence microscopy to detect apoptotic cells. The Annexin A5 affinity assay typically involves conjugating Annexin A5 with a fluorescent or enzymatic label, allowing for the distinction between viable, apoptotic, and necrotic cells . This assay is widely used in research to study cell death mechanisms and evaluate the efficacy of therapeutic agents.