FAS (TNFRSF6) is a type I transmembrane protein with an extracellular ligand-binding domain and an intracellular "death domain" critical for apoptosis signaling . FAS antibodies bind to this receptor, mimicking or blocking its natural ligand, FAS ligand (FASL).
Key Mechanisms:
Agonist Antibodies: Trigger apoptosis by clustering FAS receptors, recruiting FADD (Fas-associated death domain) and caspase-8 to form the death-inducing signaling complex (DISC) .
Antagonist Antibodies: Inhibit FASL binding, preventing apoptosis and modulating immune responses .
Designed to induce apoptosis in target cells (e.g., cancer cells). Examples include:
Block FAS-FASL interactions to prevent apoptosis. Example: PC111, a human anti-FASL mAb, inhibits blister formation in pemphigus by neutralizing soluble FASL .
Cancer Therapy: Agonist antibodies (e.g., E09) induce tumor cell apoptosis but face challenges like hepatotoxicity (e.g., Jo2) .
Autoimmune Diseases: PC111 reduces acantholysis in pemphigus by blocking FASL-mediated keratinocyte apoptosis .
Affinity Paradox: Higher-affinity FAS antibodies (e.g., EP6b_B01) show reduced agonism due to slower dissociation rates, limiting receptor clustering .
FcγR Dependence: Antibody cytotoxicity varies with Fcγ receptor interactions. For example, Jo2 requires FcγRIIB for hepatotoxicity, while HFE7A (non-toxic) relies on FcγRI/III .
Hepatotoxicity: Systemic FAS activation via antibodies like Jo2 causes fatal liver damage .
Dosing Complexity: Optimal activity requires balancing affinity, valency, and FcγR interactions .
FAS (also known as CD95, APT1, TNFRSF6) functions as a receptor for TNFSF6/FASLG. Upon binding with its ligand, FAS recruits the adapter molecule FADD, which in turn recruits caspase-8 (CASP8) to form the death-inducing signaling complex (DISC). This complex initiates a cascade of caspase activation that ultimately leads to programmed cell death or apoptosis . Beyond its role in general apoptosis, FAS-mediated cell death serves specific immunological functions, including peripheral tolerance induction and antigen-stimulated suicide of mature T-cells . This mechanism helps maintain immune homeostasis by eliminating activated lymphocytes after immune responses and removing potentially autoreactive cells.
FAS antibodies demonstrate varying capabilities to induce apoptosis, which can be quantified through efficiency (maximal cell killing percentage) and potency (EC50). Research comparing different antibodies with FAS ligand (FasL) reveals significant variations in agonistic activity. For example, the fully human anti-FAS antibody E09 demonstrates an efficiency of approximately 80% with an EC50 of 0.9 nM, while the natural ligand FasL shows 94% efficiency with an EC50 of 7 nM . Other agonistic antibodies like DX2 and SM1.1 demonstrate reduced cell-killing efficiencies of 16% and 26%, respectively . These differences appear not to be based simply on affinity, as Kd values for these antibodies fall within a similar range (5-17 nM) . Rather, epitope recognition and binding kinetics play crucial roles in determining agonistic potential.
FAS antibody-induced apoptosis triggers a series of detectable cellular changes that can be measured by various experimental techniques. Within 5 hours of stimulation with immobilized anti-FAS antibody, enhanced DNA fragmentation (a hallmark of apoptosis) can be observed in vulnerable cells . Flow cytometry using Hoechst 33342 (HO342) staining reveals morphological changes associated with apoptosis, and these cells typically double-stain with Merocyanine 540, another apoptotic indicator . Additionally, hypodiploid nuclei can be detected when stained with hypotonic propidium iodide solution. The apoptotic process ultimately leads to a reduction in the number of viable cells recovered from culture . These changes affect both CD4+ and CD8+ T cell populations, though preliminary evidence suggests CD8+ T cells may undergo apoptosis with faster kinetics than CD4+ T cells .
When selecting an anti-FAS antibody for research, consider multiple factors beyond simple reactivity to the target. First, determine the experimental application (Western blotting, immunoprecipitation, functional studies) as antibodies often perform differently across applications. For example, the rabbit polyclonal Fas antibody (ab82419) is suitable for Western blotting and reacts with rat and mouse samples . Second, consider the epitope recognition properties, as antibodies targeting different epitopes can produce dramatically different biological effects. The search results show that while several antibodies (E09, SM1.1, DX2) all competed with FasL binding, they did not all compete with each other, indicating different binding sites on the receptor . Third, examine binding kinetics, which critically impacts functional outcomes. Antibodies with faster off-rates (like E09) trigger caspase cascades more rapidly than those with slower dissociation rates . Finally, verify species reactivity to ensure the antibody recognizes your experimental model organism.
Antibody | Species Reactivity | Applications | Efficiency in Apoptosis | EC50 |
---|---|---|---|---|
ab82419 (Rabbit Polyclonal) | Mouse, Rat | WB | Not specified | Not specified |
E09 (Human IgG1) | Human | Functional assays | 80% | 0.9 nM |
DX2 (Mouse) | Human | Functional assays | 16% | Not specified |
SM1.1 (Mouse) | Human | Functional assays | 26% | Not specified |
FAS-mediated apoptosis can be measured through multiple complementary techniques that assess different stages of the apoptotic process. For early apoptosis detection, measure caspase 3/7 activation, which indicates the initiation of the apoptotic cascade . Flow cytometry using Hoechst 33342 staining provides quantitative assessment of apoptotic cells, with additional confirmation possible through Merocyanine 540 double-staining . DNA fragmentation analysis serves as another definitive method, detectable within 5 hours of anti-Fas stimulation in sensitive cells . For later apoptotic events, measure hypodiploid nuclei using hypotonic propidium iodide solution. Cell viability assays can determine both the efficiency (maximal cell killing percentage) and EC50 (concentration for half-maximal effect) of anti-FAS antibodies . When conducting these experiments, always include appropriate controls: untreated cells for baseline apoptosis levels and isotype control antibodies to confirm specificity of the anti-FAS effect . The experimental design should also account for kinetic differences, as some cell populations (like CD8+ T cells) may undergo apoptosis more rapidly than others (CD4+ T cells) .
Robust experimental design for studying FAS receptor signaling requires several essential controls. First, include untreated cells to establish baseline levels of spontaneous apoptosis, which may vary between different cell types and disease states . Second, incorporate isotype-matched control antibodies to distinguish specific FAS-mediated effects from non-specific antibody binding effects. Research shows that isotype control antibody-treated cultures typically do not differ from spontaneously occurring apoptosis . Third, when comparing efficacy of different FAS antibodies, include the natural ligand FasL as a physiologically relevant reference point . For mechanistic studies, consider antibodies targeting other TNF receptor family members as negative controls; research demonstrates that antibodies to CD27, CD30, CD40, OX40, 4-1BB, p55 TNF receptor, and p75 TNF receptor failed to induce T cell apoptosis above background levels in contexts where anti-FAS antibodies were effective . Finally, when studying kinetics, collect data at multiple time points to capture the full progression of the apoptotic process, as different cell populations may exhibit distinct temporal responses to FAS stimulation .
Counter-intuitively, higher affinity does not correlate with increased agonistic activity in anti-FAS antibodies. Research demonstrates a negative correlation between Fas affinity and cell-killing efficiency . When the E09 antibody was affinity-matured through directed evolution, the highest-affinity variant (EP6b_B01) completely lost its apoptosis-inducing ability despite binding more strongly to the receptor . Intermediate affinity-optimized variants showed reduced efficiency compared to the original E09 antibody (43% versus 75%) . This phenomenon was confirmed through rational point mutations of E09 designed to modulate Fas binding affinity.
The mechanism behind this counterintuitive relationship appears to involve binding kinetics, particularly the dissociation rate. Time-course experiments measuring caspase 3/7 activation revealed that antibodies with faster off-rates (like E09) triggered apoptotic cascades more rapidly than their higher-affinity, slower-dissociating counterparts . This suggests that optimal receptor clustering and subsequent signal transduction may require dynamic binding and unbinding events rather than stable, high-affinity interactions. These findings have significant implications for therapeutic antibody development, challenging the conventional assumption that higher affinity necessarily leads to improved biological activity.
The structural basis of FAS epitope recognition reveals important insights into antibody function. Crystallographic studies of the E09:Fas complex at 1.9 Å resolution demonstrate that agonistic antibodies do not necessarily mimic the natural ligand's binding mode . Of the residues crucial for FasL binding, only two (Fas_81F and Fas_86R) were found in the antibody epitope . When FasL was modeled onto the structure, only one loop of FasL overlapped with a loop of the agonistic antibody—specifically the DE loop containing the highly conserved XYP motif critical for death ligand-receptor interactions .
Cell-type specific variations in FAS-mediated apoptosis sensitivity represent an important consideration in experimental design. Research comparing HIV-infected and uninfected individuals demonstrates dramatic differences in cellular susceptibility to anti-FAS antibody-induced apoptosis. T lymphocytes from HIV-infected individuals exhibit markedly higher levels of apoptosis when stimulated with anti-FAS antibody compared to cells from uninfected controls . Within the T cell compartment, both CD4+ and CD8+ subpopulations undergo apoptosis, though preliminary evidence suggests CD8+ T cells may have earlier kinetics of apoptosis induction .
The data indicate substantial variability in apoptotic response based on disease state:
Cell Population | HIV+ Individuals (n=79) | HIV- Individuals (n=16) |
---|---|---|
CD3+ lymphocytes | 31 ± 2% | 7 ± 2% |
CD4+ lymphocytes | 27 ± 3% | Not specified |
CD8+ lymphocytes | 38 ± 3% | Not specified |
Further stratification shows that symptomatic HIV+ individuals (n=31) exhibited higher apoptosis levels (45 ± 3%) compared to asymptomatic individuals (n=48) with 17 ± 2% . These variations highlight the importance of considering pathological state when studying FAS-mediated apoptosis and selecting appropriate experimental controls.
Variable responses to anti-FAS antibody treatment can stem from multiple factors that researchers should systematically address. First, antibody format critically impacts functionality—solid-phase (plastic-bound) anti-FAS antibodies induce more robust apoptosis than soluble antibodies, as demonstrated in studies with the M3 antibody . Second, the binding kinetics of the antibody significantly influence biological outcomes; antibodies with similar affinities but different on/off rates produce dramatically different apoptotic responses . Third, target cell susceptibility varies substantially based on activation state, disease context, and cell type. For instance, T cells from HIV-infected individuals demonstrate markedly higher susceptibility to FAS-mediated apoptosis compared to healthy controls .
Additionally, experimental timing affects outcomes, as apoptotic cascades progress through distinct phases with different measurable endpoints. Early apoptosis (caspase activation) and late apoptosis (DNA fragmentation) measurements may yield different results depending on the timepoint examined . Finally, consider cross-reactivity with other TNF receptor family members or non-specific antibody effects by always including appropriate isotype controls and specificity verification . Standardizing these variables across experiments will improve reproducibility and accurate interpretation of FAS-mediated apoptosis studies.
Distinguishing FAS-mediated apoptosis from other cell death mechanisms requires a multi-parameter approach. First, validate pathway specificity by comparing anti-FAS antibody-induced apoptosis with control antibodies targeting other TNF receptor family members. Research demonstrates that while anti-FAS antibodies induce significant apoptosis, antibodies to CD27, CD30, CD40, OX40, 4-1BB, p55 TNF receptor, and p75 TNF receptor fail to induce T cell apoptosis above background levels , confirming pathway specificity.
Second, monitor the sequential hallmarks of FAS-mediated apoptosis: early caspase 3/7 activation followed by DNA fragmentation and membrane changes. FAS signaling specifically activates the extrinsic apoptotic pathway through FADD recruitment and caspase-8 activation . Third, use pathway inhibitors strategically—caspase inhibitors should block FAS-mediated apoptosis if the canonical pathway is engaged. Fourth, examine morphological changes distinctive to apoptosis versus necrosis or pyroptosis using microscopy and flow cytometry with Hoechst 33342 and Merocyanine 540 double-staining .
Finally, consider kinetic analysis, as genuine FAS-mediated apoptosis shows characteristic temporal progression in caspase activation . Together, these approaches provide confidence in identifying authentic FAS-specific cell death versus other pathways or non-specific effects.
The unexpected inverse relationship between anti-FAS antibody affinity and agonistic activity represents a fascinating paradox with important implications for therapeutic antibody development. Higher-affinity anti-FAS antibodies consistently demonstrate reduced apoptosis-inducing capability, contrary to conventional expectations . This phenomenon can be explained by a model that prioritizes binding kinetics over equilibrium affinity. Antibodies with faster dissociation rates (koff) more effectively trigger apoptosis, suggesting that dynamic binding and unbinding events are crucial for optimal receptor clustering and subsequent signal transduction .
Time-course experiments measuring caspase 3/7 activation confirm this hypothesis, showing that antibodies with faster off-rates (like E09) triggered apoptotic cascades more rapidly than their higher-affinity, slower-dissociating counterparts . This kinetic proofreading model suggests that receptor activation may require a specific temporal pattern of engagement rather than stable, persistent binding. The findings parallel observations in T cell receptor signaling, where serial engagement of multiple receptors by a single ligand proves more effective than stable binding.
For researchers developing agonistic antibodies, these results highlight the importance of optimizing binding kinetics rather than pursuing maximum affinity. When encountering contradictory activity data, examining dissociation rates may provide mechanistic insights into varying biological outcomes.
Structural studies of FAS-antibody complexes provide valuable templates for therapeutic agonist development. Crystallographic analysis of the E09:Fas complex at 1.9 Å resolution revealed that effective agonist antibodies need not mimic the entire binding interface of the natural ligand . Instead, key interaction points appear sufficient for triggering receptor activation. The overlap of antibody VH Y100d with FasL Y218 at a critical receptor interaction site demonstrates how partial structural mimicry can achieve similar functional outcomes .
Future research should explore minimal epitope requirements for receptor activation and identify critical "hotspots" that determine agonistic potential. Structure-guided engineering could then optimize these interaction sites while maintaining appropriate binding kinetics. The negative correlation between affinity and agonistic activity suggests that designed therapeutics should prioritize optimal binding dynamics over maximum affinity . Additionally, investigating the structural basis of receptor clustering and DISC formation following antibody binding could inform the development of antibodies with enhanced signaling capabilities.
Comparative analysis of antibodies with varying agonistic potentials provides an opportunity to establish structure-activity relationships that could guide rational design approaches. Ultimately, integrating structural insights with functional assays and binding kinetics will accelerate the development of next-generation therapeutic agonists with improved specificity and efficacy.
Advancing our understanding of FAS signaling across disease contexts requires methodological innovations addressing current technical limitations. Single-cell analysis technologies would help decipher the heterogeneity in FAS expression and response within complex tissues, moving beyond population-averaged measurements that mask important cellular subsets. Time-resolved imaging techniques could visualize the dynamic process of receptor clustering, DISC formation, and subsequent signaling events in living cells, providing insights into the temporal regulation of FAS-mediated apoptosis.
Disease-relevant model systems are needed to accurately recapitulate pathological conditions. The dramatic differences in FAS sensitivity between HIV-infected and healthy individuals highlight the importance of appropriate disease models . Developing organoid systems or humanized mouse models that faithfully reproduce disease-specific FAS signaling abnormalities would advance translational research.
Standardized quantification methods would improve comparability across studies. Currently, apoptosis measurements vary (caspase activation, DNA fragmentation, membrane changes), making direct comparisons challenging. A consensus panel of measurements with standardized protocols would facilitate more robust meta-analyses. Finally, integrated multi-omics approaches combining proteomics, transcriptomics, and phospho-signaling data could provide comprehensive pathway maps revealing how FAS signaling interfaces with other cellular processes in health and disease, potentially identifying new therapeutic targets or biomarkers.
The FAS Blocking/Activating Antibody, also known as CD95, is a monoclonal antibody derived from mice and is specifically designed to target human FAS (CD95) proteins. This antibody plays a crucial role in the study of apoptosis, a form of programmed cell death, by either blocking or activating the FAS receptor.
FAS, also known as CD95 or APO-1, is a cell surface receptor that belongs to the tumor necrosis factor receptor superfamily. It is widely expressed on the surface of various cell types, including both normal and transformed cells. The primary function of FAS is to mediate apoptosis when it binds to its ligand, FASL (CD95L/Apo-1L). This interaction triggers a cascade of intracellular signaling events leading to cell death .
The FAS Blocking/Activating Antibody can function in two distinct ways:
The FAS Blocking/Activating Antibody is utilized in a variety of research applications, including:
The antibody is typically shipped in a lyophilized form at ambient temperature. For long-term storage, it should be kept at -20°C. After reconstitution with water, it is recommended to aliquot and store the antibody at -20°C if not used within a month. Proper storage ensures the stability and efficacy of the antibody for up to 12 months .