Fact-checked by Grok 2 weeks ago

Fas receptor

The Fas receptor, also known as CD95 or APO-1, is a type I encoded by the and belonging to the (TNF) receptor superfamily, which mediates or upon binding to its ligand, (FasL). This receptor is widely expressed on the surface of various cell types, particularly in the , where it plays a pivotal role in regulating cell survival and death to maintain . Structurally, the Fas receptor features an extracellular domain with cysteine-rich repeats for ligand binding, a transmembrane region, and an intracellular death domain (DD) that is essential for signal transduction. Upon FasL engagement, the receptor trimerizes, recruiting the adaptor protein FADD and initiator caspase-8 to form the death-inducing signaling complex (DISC), which activates downstream effector caspases (such as caspases 3, 6, and 7) to execute apoptosis. In certain cells like hepatocytes, this pathway is amplified via the mitochondrial route involving Bid cleavage, whereas in lymphocytes, it can proceed directly without this amplification. In the , Fas receptor signaling is critical for eliminating pathogen-infected cells, autoreactive lymphocytes, and activated immune cells after an , thereby preventing and controlling tumor surveillance. Dysregulation of this pathway, often due to gene mutations, is associated with (ALPS), characterized by lymphoproliferation, , and increased risk, as well as contributions to conditions like and certain cancers (e.g., , , and esophageal) through impaired .

Genetics

FAS Gene

The FAS gene, located on the long arm of human at cytogenetic band 10q23.31 (chr10:88,953,813-89,029,605, GRCh38.p14), spans approximately 76 kilobases of genomic DNA and consists of nine exons that encode the precursor protein for the receptor. The gene's structure includes a TATA-less promoter region with multiple transcription initiation sites, and its introns vary significantly in length, ranging from less than 1 kilobase to over 14 kilobases, particularly at the 5' end; intron-exon splice junctions conform to the standard GT-AG consensus rule. Orthologs of the gene are widely conserved across mammals, with the mouse counterpart (Fas) mapped to , reflecting shared evolutionary origins in apoptotic regulation. A notable feature of the FAS gene structure is the evolutionary conservation of exon 9, which encodes the intracellular death domain critical for ; this exon shows high sequence similarity across vertebrates, underscoring its functional importance. Germline mutations in the FAS gene, predominantly heterozygous variants acting in a dominant-negative manner—often within the death domain-encoding exon 9—disrupt receptor function and cause (ALPS) type Ia, characterized by defective lymphocyte and lymphoproliferation. Alternative splicing of the FAS pre-mRNA generates multiple isoforms, including a soluble variant lacking the transmembrane domain encoded by exon 6.

Expression and Regulation

The FAS gene displays distinct tissue-specific expression patterns that contribute to its role in immune homeostasis and cell death regulation. Fas receptor protein is highly expressed in activated lymphocytes, where it facilitates the elimination of autoreactive or excess immune cells, as well as in the liver and thymus, organs critical for immune surveillance and T-cell maturation. In contrast, basal expression of Fas is low in neurons, rendering them relatively resistant to Fas-mediated apoptosis under normal conditions, though inducible expression can occur during pathological stress. This differential expression underscores Fas's selective involvement in immune rather than neuronal tissues. Regulation of FAS transcription occurs through multiple mechanisms, including key transcription factors and epigenetic modifications. The transcription factor NF-κB directly binds to sites in the Fas promoter, promoting its expression in response to inflammatory signals and thereby enhancing susceptibility to apoptosis in immune cells. Similarly, p53 acts as a tumor suppressor by upregulating Fas transcription following DNA damage, which sensitizes cells to death receptor signaling. Epigenetically, hypermethylation of the Fas promoter CpG islands frequently silences gene expression in various cancers, such as colon carcinoma, allowing tumor cells to evade apoptosis; this methylation pattern is observed in approximately 50% of examined colon cancer cell lines. MicroRNA-mediated post-transcriptional regulation also modulates Fas levels, with examples like miR-21 contributing to suppression of Fas-related apoptotic pathways in certain contexts, such as cardiomyocytes, by targeting upstream regulators. Post-transcriptional control further diversifies function via , which generates at least eight transcript variants in humans, including isoforms subject to . A prominent variant skips 6, encoding the , to produce a soluble isoform that circulates as a decoy receptor, binding and inhibiting in target cells. This soluble form is elevated in conditions like cancer and autoimmune diseases, where it promotes immune evasion. Environmental factors dynamically influence expression, particularly through signaling and responses. Interferon-γ (IFN-γ), a proinflammatory , upregulates Fas surface expression on various cell types, including cells, enhancing their sensitivity to Fas ligand-induced death. Other cytokines, such as TNF-α, synergize with IFN-γ to induce Fas in and endothelial cells. signals, including DNA-damaging agents like UV or chemotherapeutic drugs, also trigger Fas upregulation via activation of transcription factors such as AP-1 and , linking environmental insults to apoptotic preparedness.

Structure

Protein Domains

The Fas receptor, also known as CD95 or TNFRSF6, is a 319-amino-acid type I transmembrane with a molecular weight of approximately 48 kDa in its mature form. This architecture positions the receptor as a key member of the tumor necrosis factor receptor superfamily, spanning the plasma membrane to facilitate extracellular . The extracellular domain consists of three cysteine-rich subdomains (CRD1, CRD2, CRD3), characterized by conserved disulfide bonds that stabilize the structure for interactions. A single transmembrane helix of 17 amino acids anchors the receptor, providing a hydrophobic segment essential for membrane integration. The intracellular domain features an 80-amino-acid death domain (DD, approximately residues 210-289), a globular motif with six alpha-helices that mediates protein-protein interactions for downstream signaling. Post-translational modifications significantly influence Fas function and stability. N-glycosylation occurs at asparagine residues 102 and 120 in the extracellular region, contributing to the glycoprotein nature and observed molecular weight variations (40-55 kDa). Palmitoylation at cysteine 199 in the cytoplasmic tail enhances membrane association and prevents lysosomal degradation. Fas exists as membrane-bound and soluble isoforms due to . The canonical membrane-bound isoform retains all domains, including the transmembrane and , for surface expression and full signaling capability.00864-8) The soluble isoform arises from skipping 6, which encodes the transmembrane region, yielding a truncated protein (approximately 277 ) comprising primarily the extracellular domain; this secreted form lacks membrane anchoring and intracellular signaling elements, altering its role in modulation.00864-8)

Oligomerization and Variants

The Fas receptor (CD95) exists in pre-ligand-associated states, forming inactive oligomers primarily as dimers or trimers through interactions involving its pre-ligand assembly domain (PLAD) in the extracellular region and the . In the classical trimerization model, these preassembled trimers maintain a conformation that prevents signaling until (FasL) binding induces a conformational shift, exposing the intracellular death domain (DD) for downstream interactions. However, updated structural predictions from 2025, using AlphaFold-Multimer, challenge the strict requirement for trimers, proposing that anti-parallel dimers represent a predominant non-signaling state where ligand-binding sites are masked, and FasL engagement drives a transition to parallel dimers or higher-order aggregates to initiate activation. This dynamic equilibrium highlights the receptor's capacity for ligand-independent basal oligomerization, with transmembrane homotrimerization further stabilizing pre-ligand clusters. Alternative splicing of the FAS gene produces at least seven protein isoforms, which exhibit diverse structural and functional properties. A prominent variant, resulting from skipping of exon 6 (Δexon6), encodes a soluble isoform lacking the transmembrane domain, rendering it incapable of membrane anchoring and signal transduction; this anti-apoptotic form acts as a decoy by sequestering FasL in the extracellular space. Other isoforms arise from splicing events in the cytoplasmic region, including those that delete portions of the DD (e.g., via exon 7 skipping), producing membrane-bound decoy receptors that inhibit apoptosis by interfering with wild-type Fas clustering without eliciting death signals. These variants modulate the receptor's sensitivity to activation, with soluble and DD-truncated forms often upregulated in contexts of immune evasion or tumor survival. Recent cryogenic electron microscopy (cryo-EM) studies have provided insights into the structural dynamics of DD clustering upon activation. In the death-inducing signaling complex (DISC), Fas DDs assemble with Fas-associated death domain (FADD) proteins into an asymmetric oligomeric network, such as a 7:5 Fas DD:FADD DD complex, where sequential type I, II, and III interactions propagate higher-order clustering to amplify signaling. This architecture reveals how ligand-induced conformational changes in the receptor promote DD exposure and FADD recruitment, forming a platform for caspase-8 activation without strict stoichiometry. Oligomerization stability is influenced by membrane microenvironments, particularly cholesterol-rich lipid rafts, which facilitate Fas partitioning and enhance receptor clustering. In these rafts, sphingolipid-cholesterol interactions concentrate Fas molecules, promoting efficient FasL-induced aggregation and DISC formation, while depletion of cholesterol disrupts this organization and attenuates signaling. Such raft association underscores the role of lipid composition in fine-tuning the receptor's oligomeric state and responsiveness.

Ligand and Activation

Fas Ligand

The (FasL), also known as CD95L or CD178, is a type II transmembrane protein belonging to the (TNF) superfamily, with a molecular weight of approximately 40 kDa.90626-L) It features an N-terminal cytoplasmic domain, a transmembrane region, and a C-terminal extracellular TNF homology domain (THD) that facilitates homotrimerization, essential for its . The trimeric structure of FasL allows it to engage multiple Fas receptors simultaneously, promoting efficient signaling. A soluble variant of FasL is generated through proteolytic cleavage by metalloproteinases, primarily at the stalk region adjacent to the transmembrane domain, resulting in a circulating form that retains trimeric configuration and receptor-binding capability. FasL expression is predominantly restricted to activated T lymphocytes and natural killer (NK) cells, where it is upregulated following immune stimulation to mediate cytotoxicity against target cells. Constitutive expression occurs in immune-privileged tissues, such as the eye and testis, contributing to the maintenance of local tolerance by eliminating infiltrating inflammatory cells. FasL interacts with the Fas receptor through its cysteine-rich domains (CRDs) with high affinity, characterized by a (Kd) in the low nanomolar range, approximately 1 , enabling specific and potent . This binding initiates Fas receptor clustering and , triggering downstream cellular responses.30299-4) Regulation of FasL involves both transcriptional and post-translational mechanisms; its expression is induced in T cells by signals such as receptor and . Shedding of the membrane-bound form to generate soluble FasL is primarily mediated by the metalloproteases ADAM10 and ADAM17, with ADAM10 responsible for constitutive release in resting cells and both contributing to enhanced shedding upon T cell via PKC and calcium-dependent pathways.

Receptor Activation Mechanism

The Fas receptor (CD95) exists on the cell surface as preassembled, inactive trimers, maintained in a conformation that sequesters its intracellular death domain (DD). Activation begins when trimeric (FasL), typically membrane-bound, engages the extracellular cysteine-rich domains of these preformed Fas trimers, inducing higher-order clustering into supramolecular signaling platforms. This ligand-induced aggregation, requiring at least two adjacent FasL trimers to effectively multiple Fas molecules, transforms the inactive complexes into active assemblies capable of signal initiation. Upon clustering, the Fas receptors undergo a critical conformational change that exposes the previously buried in the cytoplasmic tail. This exposure enables homotypic - interactions with the adaptor protein (Fas-associated death domain protein), recruiting it to the receptor complex and positioning FADD's death effector domain for subsequent interactions. The structural basis of this DD activation involves a rotation and opening of the DD helix bundle, relieving autoinhibitory constraints and facilitating adaptor binding without altering the overall trimer symmetry. Effective activation demands a level of receptor and clustering, typically involving multiple FasL molecules per receptor to achieve sufficient and stability. localization of FasL is essential, as it promotes dense, planar interactions that drive robust clustering, whereas soluble FasL often fails to induce signaling due to its inability to mimic this geometry and instead acts as a competitive . Cellular factors, such as receptor density and lateral mobility within the plasma , further modulate this by influencing clustering . Inhibitory mechanisms counteract activation to prevent inappropriate signaling. The soluble decoy receptor DcR3 (decoy receptor 3) binds FasL with high affinity, sequestering it and preventing productive engagement with membrane Fas receptors. Similarly, soluble forms of FasL or Fas itself can interfere by competing for binding sites or disrupting cluster formation, thereby raising the activation threshold in physiological contexts.

Signaling Pathways

Apoptotic Pathway

Upon trimerization of the Fas receptor induced by binding, the intracellular death domains (DDs) of Fas recruit the adaptor protein through homotypic DD-DD interactions. This recruitment occurs rapidly at the plasma membrane, forming the core of the death-inducing signaling complex (DISC). FADD, in turn, binds procaspase-8 via death effector domain (DED)-DED interactions between the DEDs of FADD and the prodomain of procaspase-8, leading to the aggregation of multiple procaspase-8 molecules within the DISC. Within the DISC, proximity-induced dimerization of procaspase-8 promotes its autoactivation through proteolytic cleavage, generating active heterotetramers. Active then initiates the caspase cascade by directly cleaving and activating downstream effector caspases, such as caspase-3 and caspase-7. In parallel, cleaves the BH3-only protein Bid at aspartate 60 in humans (or 59 in mice), producing truncated Bid (tBid), which translocates to mitochondria and amplifies the apoptotic signal by activating the intrinsic pathway through Bax/Bak oligomerization and release. This crosstalk between extrinsic and intrinsic pathways enhances the efficiency of in certain cell types. The execution phase of Fas-mediated involves the proteolytic activity of effector , leading to the dismantling of cellular structures. Caspase-3 and caspase-7 cleave key substrates, including PARP, , and ICAD, resulting in DNA fragmentation via caspase-activated DNase (CAD), condensation, and nuclear breakdown.55055-5/fulltext) Cytoskeletal rearrangements driven by caspase-mediated cleavage of and contribute to cell shrinkage and membrane blebbing, hallmark morphological features of .55055-5/fulltext) These events culminate in the formation of apoptotic bodies, which are phagocytosed without eliciting . Fas signaling exhibits cell-type specificity, classified into Type I and Type II cells based on the reliance on the direct versus mitochondrial amplification. In Type I cells, such as thymocytes, high levels of formation lead to robust activation sufficient to directly trigger effector without significant mitochondrial involvement. Conversely, in Type II cells, like hepatocytes, weaker signaling results in limited initial activity, necessitating tBid-mediated mitochondrial amplification to achieve full effector activation and . This distinction influences sensitivity to Fas-mediated death and therapeutic targeting in diseases.

Non-Apoptotic Pathways

In addition to its role in , the Fas receptor (CD95) engages non-apoptotic signaling pathways that promote cell survival, proliferation, and inflammatory responses. These pathways often share initial components of the death-inducing signaling complex (), such as , but diverge to activate pro-survival cascades depending on cellular context and adaptor proteins. One prominent non-apoptotic pathway involves Fas-induced activation of the transcription factor, mediated by receptor-interacting protein kinase 1 () and transforming growth factor-β-activated kinase 1 (TAK1). Upon Fas ligation, recruits TAK1 to the signaling complex, where TAK1 phosphorylates the (IKK) complex, leading to IκB degradation and nuclear translocation of . This results in the transcription of anti-apoptotic genes such as and cIAPs, thereby inhibiting cell death and promoting survival in various cell types, including immune and epithelial cells. Fas signaling also activates the (MAPK)/extracellular signal-regulated kinase (ERK) pathway, particularly in cancer cells, where it enhances and invasion. In contexts of impaired apoptotic signaling, such as Fas mutations, ERK activation drives cytoskeletal remodeling and expression, facilitating metastatic potential; for instance, in oral squamous cell carcinoma, Fas engagement promotes ERK-dependent JAG1 upregulation, supporting epithelial-mesenchymal transition and invasive behavior. In B lymphocytes, non-apoptotic Fas signaling modulates the mechanistic target of rapamycin () pathway, influencing metabolic responses and extrafollicular maturation. Fas ligation regulates PTEN nuclear exclusion, thereby enhancing CD40-induced PI3K/AKT/ activation, which supports proliferation and production without triggering death; this mechanism is critical for rapid in humans. Furthermore, Fas contributes to inflammatory responses by stimulating cytokine production and immune cell activation, aiding clearance. In myeloid cells and during infections, Fas promotes and MAPK-dependent secretion of pro-inflammatory cytokines like TNF-α and IL-6, enhancing and activation for efficient elimination of pathogens such as and viruses.

Protein Interactions

Key Interactors

The Fas receptor (also known as CD95) primarily interacts with adaptor proteins through its intracellular (DD), facilitating the recruitment of downstream effectors in and non-apoptotic signaling. The key adaptor (Fas-associated death domain protein) binds directly to the Fas DD via a homotypic DD-DD interaction, which is essential for assembling the -inducing signaling complex () and initiating activation. This interaction occurs upon Fas trimerization induced by binding, positioning FADD's death effector domain (DED) to recruit other components. Another adaptor, Daxx (death domain-associated protein), serves as an alternative DD binder to Fas, independent of FADD, and promotes a distinct signaling axis involving JNK activation rather than -dependent . Daxx engages a specific region within the Fas DD, enabling its translocation from the to the upon receptor activation. Caspases and their regulators interact indirectly with Fas through FADD but are critical components of the primary signaling hub. Procaspase-8 and procaspase-10 bind to FADD's DED via their own DEDs in a homotypic manner, forming oligomers that promote autoactivation and apoptotic signal propagation. The inhibitory homolog c-FLIP (cellular FLICE-like inhibitory protein) competes for the same DED-binding sites on FADD as procaspase-8, thereby modulating assembly and suppressing while favoring non-apoptotic outcomes like activation. Additional interactors include proteins involved in non-apoptotic pathways and post-translational modifications. The BH3-only protein BID engages indirectly following its cleavage by activated , with the resulting truncated BID (tBID) amplifying mitochondrial apoptosis but not binding Fas directly. (receptor-interacting serine/threonine kinase 1) binds directly to the Fas DD, contributing to non-apoptotic signaling such as necroptosis and by competing with FADD for binding sites and forming oligomeric complexes. Src family kinases, particularly , mediate tyrosine within the Fas death domain, enhancing non-apoptotic survival signals and receptor desensitization. Quantitative analyses of Fas interactions reveal defined stoichiometries in signaling complexes, underscoring their in cellular . For instance, in the , DED-containing proteins like procaspase-8 and c-FLIP outnumber by several-fold, supporting chain-like oligomerization for efficient activation. Recent 2025 structural modeling of CD95 trimers highlights a 7:3 stoichiometry of to in necrosome-like assemblies, informing precision medicine strategies for modulating Fas signaling in immune disorders. These interactions collectively contribute to the formation of larger multi-protein complexes that dictate cellular fate.

Complex Formation

The death-inducing signaling complex (DISC) forms the core multi-protein assembly for Fas-mediated apoptosis, comprising the Fas receptor, the adaptor protein FADD, and procaspases-8 and -10, with the stoichiometry varying based on cellular context and typically involving multiple Fas trimers oligomerized via death domain interactions to recruit 3–6 FADD molecules and associated procaspases. c-FLIP integrates into the DISC as a regulatory component, forming heterodimers with caspase-8 to inhibit its activation and thereby modulate apoptotic output through variable ratios that can range from pro-apoptotic (low FLIP) to anti-apoptotic (high FLIP) configurations. Beyond the canonical , assembles alternative complexes that drive non-apoptotic signaling, such as the Fas-Daxx complex, where Daxx binds directly to the Fas death domain to activate the JNK pathway independently of . In other contexts, incorporates alongside and to form complexes that promote activation, particularly when caspase-8 activity is limited, enabling survival or inflammatory responses. The dynamics of Fas complex assembly are rapid, with DISC formation occurring within seconds of receptor trimerization, driven by sequential of FADD and procaspases to achieve functional oligomerization. Disassembly follows through ubiquitination of key components like FADD by E3 ligases such as MKRN1, targeting them for proteasomal degradation and terminating signaling. Complex stability is further modulated by localization to rafts, which facilitate Fas clustering and enhance DISC efficiency, while events, such as Cdk1-mediated modification of procaspase-8, alter and thresholds within the complex.

Biological Roles

In Immune Regulation

The Fas receptor plays a pivotal role in immune regulation by controlling numbers and preventing aberrant immune responses through induction and, in some contexts, non-apoptotic signaling. In the , Fas-FasL interactions maintain by eliminating excess or potentially autoreactive s, thereby averting and ensuring effective clearance of pathogens or transformed cells. One key mechanism is activation-induced cell death (AICD), which curbs T cell expansion following antigen-driven activation to prevent . Upon (TCR) restimulation, activated T cells upregulate (FasL), leading to autocrine or paracrine Fas engagement that forms the death-inducing signaling complex (DISC) with and procaspase-8, initiating caspase-dependent . This process is essential for terminating immune responses, as evidenced by Fas-deficient lpr mice, which exhibit massive and systemic due to defective AICD and accumulation of autoreactive T cells. Fas also facilitates the cytotoxic activity of CD8+ cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, enabling targeted killing of virally infected or malignant cells expressing Fas. These effectors release membrane-bound FasL, which trimerizes Fas on target cells, activating the DISC and extrinsic apoptosis pathway independently of or alongside perforin/granzyme exocytosis. This FasL-mediated pathway accounts for a significant portion of CTL- and NK-induced cytotoxicity, particularly against Fas-sensitive targets, ensuring efficient immune surveillance without excessive inflammation. In , Fas contributes to the deletion of self-reactive s, maintaining immune self-tolerance beyond central mechanisms. Activated self-reactive T and B cells in peripheral tissues are eliminated via Fas-FasL interactions, often in collaboration with intrinsic pathways like Bim-mediated regulation; for instance, Fas ligation on B cells in germinal centers by T cell-derived FasL promotes of low-affinity or autoreactive clones when survival signals such as CD40 are absent. in Fas, as seen in lpr mice, disrupt this deletion, leading to unchecked self-reactive proliferation and autoimmune disorders like lupus-like syndrome. Beyond , Fas elicits non-apoptotic signals in B cells that modulate , , and production, particularly through integration with (BCR) and co-stimulatory pathways. In activated B cells, Fas engagement, combined with CD40 stimulation, activates via REL family members, upregulating anti-apoptotic c-FLIP to favor survival and over death, thereby supporting . Recent studies further reveal that non-apoptotic Fas signaling inhibits activation via caspase-8-mediated PTEN nuclear exclusion, steering B cells toward maturation and memory formation rather than extrafollicular responses; defective Fas signaling in (ALPS) patients enhances PI3K/ activity, promoting extrafollicular B cell expansion and hypergammaglobulinemia.

In Tissue Homeostasis and Development

The Fas receptor plays a critical role in maintaining liver homeostasis by regulating hepatocyte turnover and apoptosis, which is essential for balancing cell proliferation during regeneration. In the liver, Fas-FasL interactions trigger caspase-dependent apoptosis in hepatocytes, facilitating the elimination of damaged or excess cells to prevent overproliferation following injury. During active liver regeneration, such as after partial hepatectomy, Fas-mediated apoptosis is temporarily delayed to prioritize proliferative signals, allowing compensatory growth while maintaining tissue integrity; this regulation involves shedding of the Fas ectodomain to limit signaling. Thus, Fas ensures controlled hepatocyte renewal, contributing to long-term liver homeostasis beyond acute repair phases. In neural development, Fas-mediated apoptosis sculpts the by eliminating excess neurons generated during , thereby refining neural circuits and preventing overcrowding. Expression of is upregulated in the developing , where it sensitizes neurons to FasL-induced death signals, promoting in post-mitotic neurons to match target innervation requirements. This apoptotic pruning is particularly prominent in regions like the and , where overproduction of neurons occurs early in embryogenesis, and Fas signaling helps achieve the precise neuronal density needed for functional connectivity. Additionally, Fas influences dynamics, with low-level activation supporting progenitor survival and without inducing full , thus integrating death and survival cues during . Beyond , Fas engages non-apoptotic pathways to promote , aiding by facilitating tissue remodeling. In human , soluble FasL activates mitogen-activated protein kinases (MAPKs) such as ERK and JNK independently of cascades, enhancing cell motility and invasion without triggering death. This signaling supports recruitment to injury sites, where it coordinates deposition and contraction during the proliferative phase of repair. Cleaved forms of FasL further amplify via ion exchanger activation, underscoring Fas's role in dynamic cellular responses essential for efficient . Fas-deficient mice, such as the lpr strain, exhibit viable embryonic and early postnatal development with no gross tissue abnormalities, indicating functional redundancy in apoptotic pathways during . These mice develop progressive and starting around 8 weeks of age due to impaired , but their overall tissue architecture remains intact, highlighting compensatory mechanisms in non-immune contexts like liver and neural tissues. This overlaps briefly with immune regulation in lymphoid organs, where Fas loss disrupts without derailing core developmental processes.

Pathophysiology

Autoimmune Diseases

Dysfunction of the Fas receptor plays a central role in (ALPS), a rare characterized by defective leading to uncontrolled proliferation and . ALPS type Ia, the most common form, results from heterozygous mutations in the , which impair Fas-mediated and cause accumulation of autoreactive lymphocytes. These mutations often occur in the death domain of Fas or affect splicing, such as the common intron 5 splice-site that disrupts proper mRNA and protein function. The condition is typically inherited in an autosomal dominant manner, with each child of an affected individual having a 50% chance of inheriting the pathogenic variant, though is incomplete, affecting less than 60% of carriers. Clinically, manifests as chronic , , and autoimmune cytopenias, such as and , due to the failure of signaling to eliminate excess lymphocytes. Diagnosis requires chronic (>6 months) non-malignant, non-infectious or , along with elevated double-negative T cells (DNTs; TCRαβ+ CD4− CD8−), defined as ≥1.5% of total lymphocytes or ≥2.5% of CD3+ T cells. is rare, with over 500 cases reported worldwide, though exact prevalence remains unknown. Beyond , Fas pathway dysregulation contributes to other autoimmune diseases. In systemic lupus erythematosus (SLE), elevated levels of soluble (sFasL) in serum are observed, particularly in active disease, which may inhibit Fas-mediated and promote lymphocyte survival, exacerbating autoimmunity. Similarly, in , non-apoptotic Fas signaling in synovial fibroblasts and macrophages drives proinflammatory production and tissue inflammation, contributing to joint destruction independent of pathways.

Role in Cancer

The Fas receptor functions as a tumor suppressor by facilitating immune-mediated of malignant cells, primarily through interactions with (FasL) expressed on cytotoxic T cells and natural killer cells. This pathway is crucial for immune surveillance, enabling the elimination of nascent tumors and preventing their progression. Loss-of-function mutations or deletions in the are frequently observed in various cancers, impairing this apoptotic response and promoting oncogenesis. For instance, in subtypes of such as nasal natural killer/, mutant Fas transcripts, including deletions affecting the and transmembrane , occur in up to 60% of cases, contributing to resistance. Similarly, somatic FAS alterations, including deep deletions, are reported in approximately 1.9% of lung cancers, often targeting the and correlating with reduced tumor cell susceptibility to immune attack. Conversely, in established tumors, can promote cancer progression through non-apoptotic signaling pathways that enhance invasiveness and . In , Fas activation triggers extracellular signal-regulated kinase (ERK) and other (MAPK) pathways, upregulating matrix metalloproteinase-2 (MMP-2) activity via and TIMP-2 modulation, thereby increasing cell motility without inducing . Additionally, tumors exploit soluble FasL (sFasL), a cleaved form secreted by cancer cells, to evade immune detection; in colon , sFasL induces in host lymphocytes, creating an immunosuppressive niche that shields tumors from cytotoxic responses. High FAS expression often serves as a prognostic indicator of adverse outcomes in certain malignancies, reflecting its shift toward pro-tumorigenic roles. Recent 2024 oncology data from patients show that co-expression of Fas and FasL on circulating tumor cells (CTCs) is present in 84.6% of CTC-positive cases and independently predicts reduced (median 9.5 months versus 13.4 months in non-co-expressors). In the , FasL expressed on cancer cells and associated vasculature actively kills infiltrating immune cells, such as + T cells, via Fas engagement, limiting anti-tumor immunity; this mechanism is prominent in , where FasL knockout reduces T cell and enhances immune cell persistence.

Therapeutic Applications

Agonists and Antagonists

Agonists of the Fas receptor primarily function by mimicking the natural ligand FasL to induce receptor trimerization and clustering, which facilitates the assembly of the and subsequent activation of , leading to . A well-characterized example is the CH11, an agonistic anti-Fas antibody that binds to the extracellular of Fas, promotes receptor aggregation, and efficiently triggers activation and cell death in sensitive cell types such as transformed lymphocytes and certain tumor cells. Recombinant FasL trimers, engineered to replicate the oligomeric structure of the native membrane-bound ligand, also serve as potent agonists by binding to Fas and inducing formation, though their efficacy is enhanced when cross-linked to form higher-order oligomers like hexamers. Antagonists counteract Fas signaling by preventing ligand-receptor interactions or disrupting downstream DISC assembly. Soluble Fas-Fc fusion proteins, which consist of the extracellular domain of Fas linked to the Fc portion of immunoglobulin G, act as decoy receptors that competitively bind soluble or membrane-bound FasL, thereby inhibiting ligand-induced clustering and without activating the receptor themselves. Small-molecule inhibitors targeting DISC assembly, such as those interfering with recruitment or activation, can block the initiation of apoptotic signaling by stabilizing inhibitory interactions within the complex or preventing procaspase-8 dimerization. Natural modulators influence Fas activity through indirect mechanisms that alter receptor expression or signaling thresholds. Cyclosporin A, an immunosuppressive calcineurin inhibitor, enhances cellular sensitivity to Fas-mediated by abolishing costimulatory signals (e.g., from ) that otherwise confer resistance to death receptor ligation. Glucocorticoids upregulate Fas receptor expression on the surface of immune cells, such as T lymphocytes, thereby increasing susceptibility to FasL-induced cell death and contributing to the resolution of inflammatory responses. These agents, along with synthetic agonists and antagonists, have been explored in preclinical and clinical settings to modulate Fas activity for therapeutic purposes.

Clinical Trials and Emerging Therapies

Clinical trials targeting the Fas receptor (CD95) in cancer have primarily explored agonistic antibodies to induce apoptosis in tumor cells, often in combination with standard therapies. A preclinical study evaluating an agonistic anti-CD95 antibody combined with radiotherapy demonstrated enhanced antitumor effects through the abscopal effect in melanoma models, showing reduced growth rates in both primary and secondary tumors compared to controls. In lymphoma, preclinical data from bispecific anti-CD20×CD95 antibodies indicate superior cytotoxicity against malignant B cells by specifically triggering CD95-mediated death, paving the way for clinical translation. Recent advancements in CAR-T cell therapy incorporate Fas ligand (FasL) engineering to potentiate bystander killing, addressing antigen heterogeneity; a 2025 study modeled over 20% antigen loss and found enhanced Fas/FasL interactions improved tumor clearance in lymphoma and solid tumors. In autoimmune diseases, therapeutic strategies focus on restoring Fas function or blocking excessive FasL activity. For autoimmune lymphoproliferative syndrome (ALPS) caused by FAS mutations, lipid nanoparticle-mediated Fas gene therapy has shown promise in preclinical models, suppressing double-negative T cells and alleviating the ALPS phenotype in mice by restoring apoptosis. Efforts to target FasL in systemic lupus erythematosus (SLE) have provided insights despite challenges; studies have highlighted elevated soluble FasL as a marker of disease activity and informed subsequent biomarker-driven strategies. Emerging therapies leverage precision approaches to mitigate off-target effects. Bispecific antibodies linking to tumor antigens, such as in B-cell malignancies, enable targeted CD95 clustering and induction on cancer cells while sparing healthy tissues, with preclinical data supporting their role in precision oncology. For , inhibitors targeting non-apoptotic signaling pathways are under investigation; disruption of interactions in hepatocytes has been shown to reduce profibrotic responses in metabolic dysfunction-associated models, suggesting potential for clinical antifibrotic applications. Key challenges in Fas-targeted therapies include from systemic agonism, as potent anti-Fas antibodies like CH11 have induced fulminant hepatic failure in preclinical models due to widespread . Additionally, biomarkers such as Fas receptor are emerging for patient selection, with low CD95 trimerization levels correlating with resistance in tumors and guiding precision medicine strategies.

References

  1. [1]
    Fas Receptor - an overview | ScienceDirect Topics
    The Fas receptor, also known as CD95 or APO-1, is a member of the TNF receptor superfamily that mediates apoptosis or programmed cell death, ...
  2. [2]
    FAS gene: MedlinePlus Genetics
    The FAS gene provides instructions for making a protein that is involved in cell signaling. Learn about this gene and related health conditions.
  3. [3]
    The many roles of FAS receptor signaling in the immune system - PMC
    FAS and FASL play critical roles in the immune system, in particular in the killing of pathogen infected target cells and the death of no longer needed, ...
  4. [4]
    Characterization of human Fas gene. Exon/intron ... - PubMed
    Feb 1, 1995 · The gene consists of nine exons and spans more than 26 kilobases of DNA. The lengths of introns vary from > 14 kilobases at the 5' end of the ...
  5. [5]
    Entry - *134637 - FAS CELL SURFACE DEATH RECEPTOR - OMIM
    ... FAS gene is located just distal to the central part of band 10q23. Watanabe ... (1992) mapped the mouse Fas gene to the distal region of chromosome 19.Missing: mammals | Show results with:mammals
  6. [6]
    FAS Haploinsufficiency Is a Common Disease Mechanism in ... - NIH
    The FAS death domain (DD), an 85-aa-long structure encoded by exon 9, is required for FAS-induced apoptosis of lymphocytes under physiological conditions (17).Missing: clarification | Show results with:clarification
  7. [7]
    Autoimmune Lymphoproliferative Syndrome - GeneReviews - NCBI
    Sep 14, 2006 · These genes include FAS (either germline or somatic pathogenic variants), CASP10, and FASLG. Up to 20% of those with clinical ALPS have not had ...
  8. [8]
    Dominant inhibition of Fas ligand-mediated apoptosis due to a ...
    Jul 2, 2007 · Previous studies indicated that most Type Ia ALPS patients carry heterozygous Fas mutations, and that the mutated Fas molecules have dominant ...
  9. [9]
    Differential expression of human Fas mRNA species upon ... - PubMed
    Sep 15, 1995 · The gene lies on the long arm of chromosome 10, consists of nine exons, and spans more than 26 kb of DNA.
  10. [10]
    TNF-Related Ligands and Their Receptors - ScienceDirect.com
    Expression of the Fas receptor is highest in activated lymphocytes, liver, heart, ovary and thymus, although Fas can be found in many other tissues [86].
  11. [11]
    Fas/FasL Contributes to HSV-1 Brain Infection and Neuroinflammation
    Aug 29, 2021 · Although Fas is normally expressed at low levels, if at all, on neurons, during physiologic stress its expression can be induced (6, 11, 23).
  12. [12]
    NF-κB Directly Regulates Fas Transcription to Modulate Fas ... - NIH
    Fas functions as a tumor suppressor, and NF-κB directly binds to multiple sites in the Fas promoter region to regulate Fas transcription.
  13. [13]
    p53 upregulates cFLIP, inhibits transcription of NF-kappaB ...
    Although p53 upregulated the death receptors Fas, TRAIL-R1 and TRAIL-R2 in this cell line, p53-induced cell death occurred without detectable caspase-8 ...
  14. [14]
    Hypermethylation of the gene promoter and enhancer region can ...
    Apr 16, 2003 · This study determined that DNA methylation contributes to the downregulation of Fas expression in 50% of colon carcinoma cell lines examined.Results · Cpg Island Methylation · Influence Of Wtp53
  15. [15]
    MiR-21 attenuates FAS-mediated cardiomyocyte apoptosis by ... - NIH
    Our study indicates that miR-21 attenuates FAS-mediated cardiomyocyte apoptosis by regulating HIPK3 expression, which could eventually have important clinical ...
  16. [16]
  17. [17]
    Mendelian randomisation identifies alternative splicing of the FAS ...
    This soluble form of FAS acts as a decoy receptor for FASL and therefore has biologically opposing actions to membrane-bound FAS, by reducing FAS-FASL ...
  18. [18]
    Genome-Wide Identification of Fas/CD95 Alternative Splicing ...
    Jan 8, 2015 · Alternative splicing of Fas/CD95 exon 6 generates either a membrane-bound receptor that promotes, or a soluble isoform that inhibits, apoptosis.
  19. [19]
    Fas/APO-1 (CD95)–Mediated Apoptosis Is Activated by Interferon-γ ...
    In this study, we have examined the possibility to increase the sensitivity to Fas-induced apoptosis by pretreatment of MM cells with interferon-γ (IFN-γ) or ...
  20. [20]
    Up-regulation of Fas (CD95) expression in tumour cells in vivo - PMC
    TNF-alpha and IFN-gamma render microglia sensitive to Fas ligand-induced apoptosis by induction of Fas expression and down-regulation of Bcl-2 and Bcl-xL.
  21. [21]
    Signaling and transcriptional control of Fas ligand gene expression
    Mar 25, 2003 · Environmental stress stimuli such as cytotoxic stress and DNA-damaging agents can trigger responses that control cellular processes including ...
  22. [22]
    CD95 Structure, Aggregation and Cell Signaling - PMC - NIH
    May 5, 2020 · CD95 (also known as Fas, Apo-1, TNFRSF6) is a 319 amino acid type I transmembrane glycoprotein (Itoh et al., 1991; Figure 1B). In the presence ...
  23. [23]
    FAS (Fas cell surface death receptor)
    Nov 1, 2014 · CD95 (also known as Fas) is a death receptor that belongs to the TNF-receptor superfamily. Expressed at the cell surface as a homotrimer.Missing: promoter intron- evolutionary
  24. [24]
  25. [25]
    FasL-Independent Activation of Fas - NCBI - NIH
    Mature Fas (Fig. 1) is a 45-kDa type I transmembrane receptor of 319 amino acids with a single transmembrane domain of 17 amino acids (from Leu-158 to Val ...Missing: architecture | Show results with:architecture
  26. [26]
    Death domain receptors and their role in cell demise - PubMed
    The intracellular portion of all these receptors contains a region, approximately 80 amino acids long, referred to as the "death domain" (DD).
  27. [27]
    Cellular Dynamics of Fas-Associated Death Domain in the ... - MDPI
    The Structural Organization of FADD. The human FADD gene is located on chromosome 11q13.3 and consists of two coding exons that encode a 22 kDa protein [37].Missing: intron- boundaries
  28. [28]
    FAS/CD95 Polyclonal Antibody (13098-1-AP)
    6–10 day deliveryThe molecular mass of native Fas is 38 kDa, the high molecular weight form (40-55 kDa) of Fas is due to glycosylation.
  29. [29]
    Palmitoylation is required for efficient Fas cell death signaling
    We demonstrate here that Fas is palmitoylated and that this post‐translational modification of the receptor is essential for the redistribution of Fas into ...
  30. [30]
    Fas palmitoylation by the palmitoyl acyltransferase DHHC7 ... - Nature
    Oct 10, 2014 · The death receptor Fas undergoes a variety of post-translational modifications including S-palmitoylation. This protein acylation has been ...
  31. [31]
  32. [32]
    CD95/Fas stoichiometry in future precision medicine - Nature
    Apr 15, 2025 · The key feature of these TNFRs is the presence of cysteine-rich domains (CRDs), which are stabilized by cysteine disulfide bonds providing a ...
  33. [33]
    Structural Basis and Functional Role of Intramembrane Trimerization ...
    Our data suggest that the structures represent the signaling-active conformation of Fas-TM, which appears to be different from the pre-ligand conformation.
  34. [34]
    Structural Basis and Functional Role of Intramembrane Trimerization ...
    Feb 18, 2016 · Trimerization of the TM Domain Is Essential for Ligand-Induced Fas Signaling. The lack of major changes in pre-ligand receptor oligomerization ...
  35. [35]
    FAS - Tumor necrosis factor receptor superfamily member 6 - UniProt
    This entry describes 7 isoforms produced by Alternative splicing. P25445-1. This isoform has been chosen as the canonical sequence. All positional information ...
  36. [36]
    A Membrane-bound Fas Decoy Receptor Expressed by Human ...
    Sep 12, 1999 · ... we found a unique form of Fas generated by alternative splicing of exon 7, resulting in a protein lacking a death domain but with an intact.<|control11|><|separator|>
  37. [37]
    Fas/Apo-1 (CD95) receptor lacking the intracytoplasmic signaling ...
    Fas/Apo-1 (CD95) receptor lacking the intracytoplasmic signaling domain protects tumor cells from Fas-mediated apoptosis · Authors · Affiliation.
  38. [38]
    Assembly and activation of the death-inducing signaling complex
    Jun 4, 2025 · Using cryogenic electron microscopy (cryo-EM), we show that Fas and FADD death domains (DDs) form an asymmetric 7:5 oligomer, which promotes ...
  39. [39]
    The Fas/FADD death domain complex structure unravels signaling ...
    The Fas/FADD DISC represents a receptor platform, which once assembled initiates the induction of programmed cell death.
  40. [40]
    An essential role for membrane rafts in the initiation of Fas/CD95 ...
    We report here that a considerable fraction of Fas is constitutively partitioned into sphingolipid‐ and cholesterol‐rich membrane rafts in mouse thymocytes as ...
  41. [41]
    Organization and Dynamics of Fas Transmembrane Domain in Raft ...
    It has been reported that ceramide induces Fas oligomerization in lipid rafts. Here, it is shown that neither Fas TMD membrane organization nor its conformation ...
  42. [42]
    Clusters of apoptotic signaling molecule-enriched rafts, CASMERs
    May 19, 2022 · The co-clustering of Fas/CD95 and rafts facilitates DISC formation, bringing procaspase-8 molecules to be bunched together in a limited membrane ...
  43. [43]
  44. [44]
    Structural principles of tumor necrosis factor superfamily signaling
    Jan 2, 2018 · We review our current understanding of the structure and geometry of TNF superfamily ligands, receptors, and their interactions.
  45. [45]
    Conversion of Membrane-bound Fas(CD95) Ligand to Its Soluble ...
    Metalloproteinase(s) present in 293 cells cleave(s) membrane-bound FasL at the Ser126–Leu127 bond, liberating a soluble form of FasL. The cleavage site of the ...
  46. [46]
    Activation‐induced cell death: The controversial role of Fas and Fas ...
    Apr 1, 2002 · Fas is expressed ubiquitously in many tissues with particularly high expression in thymus, liver heart and kidney, whereas FasL expression is ...
  47. [47]
    The Fas signalling pathway and its role in the pathogenesis of cancer
    FasL has since been found to be expressed in sites such as the eye [2] and testis [3], where it contributes to immune privilege by inducing apoptosis of ...
  48. [48]
    Soluble Fas ligand, soluble Fas receptor, and decoy receptor 3 as ...
    DcR3 binds specifically to FasL-ECD with an affinity comparable to that of the Fas receptor (Kd = 0.8 nM) [2]. sFas, sFasL, and DcR3 ordinarily function as ...
  49. [49]
    Fas (CD95/Apo-1) ligand regulation in T cell homeostasis, cell ...
    In particular, Fas ligand (FasL), expressed by activated T lymphocytes, induces cell-mediated cytotoxicity and may also be responsible for apoptotic suicide.<|separator|>
  50. [50]
    ADAM10 regulates FasL cell surface expression and modulates ...
    Feb 9, 2007 · In primary human T cells, FasL shedding is significantly reduced after inhibition of ADAM10. The resulting elevated FasL surface expression is ...
  51. [51]
    Subcellular localization and activation of ADAM proteases ... - PubMed
    Mar 5, 2015 · In T lymphocytes, ADAM10 sheds the death factor Fas Ligand (FasL) and thereby regulates T cell activation, death and effector function.
  52. [52]
  53. [53]
    Fas death receptor signalling: roles of Bid and XIAP - PMC
    Sep 30, 2011 · Activation of Fas by Fas ligand (FasL) can induce apoptosis by a mechanism involving the adaptor protein FADD (Fas-associating protein with a ...
  54. [54]
  55. [55]
    Cytotoxicity‐dependent APO‐1 (Fas/CD95)‐associated proteins ...
    Cytotoxicity‐dependent APO‐1 (Fas/CD95)‐associated proteins form a death‐inducing signaling complex (DISC) with the receptor. F. C. Kischkel, S. Hellbardt, I.
  56. [56]
    Two CD95 (APO‐1/Fas) signaling pathways | The EMBO Journal
    We have identified two cell types, each using almost exclusively one of two different CD95 (APO‐1/Fas) signaling pathways. In type I cells, caspase‐8 was ...Results · Discussion · Materials And Methods<|control11|><|separator|>
  57. [57]
    FAS mediates apoptosis, inflammation, and treatment of pathogen ...
    Apr 22, 2025 · FAS is activated by its homologous ligand, FASL, located on the plasma membrane, which promotes aggregation and triggers conformational changes.<|control11|><|separator|>
  58. [58]
    TAK1 at the crossroads of multiple regulated cell death pathways ...
    Mar 6, 2025 · Phosphorylated TAK1 (p-TAK1) triggers the IKK complex, initiating the activation of the NF-κB pathway, which in turn regulates anti-apoptotic ...Tak1 Activation And Its... · Tak1 In The Tnf Signaling... · Regulation Of Ptms Of The...
  59. [59]
    FAS receptor regulates NOTCH activity through ERK-JAG1 axis ...
    Mar 5, 2022 · FAS knockout inhibited in vitro cancer spheroid formation, migration and invasion, and prevented mesenchymal transition in OSCC cells and ...
  60. [60]
    Non-apoptotic FAS signaling controls mTOR activation and ...
    Jan 12, 2024 · We demonstrated that FAS signaling can modulate CD40-induced mTOR activation in healthy naïve B cells by regulating PTEN nuclear exclusion and ...
  61. [61]
    Oligomerised RIPK1 is the main core component of the CD95 necrosome | The EMBO Journal
    ### Summary of Fas Trimerization and RIPK1 in CD95 Complex (2025 Insights)
  62. [62]
    Daxx, a novel Fas-binding protein that activates JNK and apoptosis
    Jun 27, 1997 · The Fas-binding domain of Daxx is a dominant-negative inhibitor of both Fas-induced apoptosis and JNK activation, while the FADD death domain ...
  63. [63]
    Human Daxx regulates Fas‐induced apoptosis from nuclear PML ...
    Daxx was first identified as a protein that binds the cytosolic domain of Fas and links this receptor to an apoptosis pathway involving activation of Jun ...
  64. [64]
    Stoichiometry of the CD95 death-inducing signaling complex
    Jul 27, 2012 · In this study we analyzed the stoichiometry of the CD95 DISC. Using quantitative western blots, mass spectrometry, and mathematical modeling, we ...
  65. [65]
    Therapeutic approaches targeting CD95L/CD95 signaling in cancer ...
    Mar 17, 2022 · Differential affinity of FLIP and procaspase 8 for FADD's DED binding surfaces regulates DISC assembly. ... Daxx, a novel Fas-binding protein ...<|control11|><|separator|>
  66. [66]
    Bid, a critical mediator for apoptosis induced by the activation of Fas ...
    Bid is a novel pro-apoptosis Bcl-2 family protein that is activated by Caspase 8 in response to Fas/TNF-R1 death receptor activation.
  67. [67]
    Membrane-bound Fas Ligand requires RIP1 for efficient activation of ...
    The serine-threonine kinase RIP1 was originally identified through its ability to bind to the death domain of Fas (CD95). RIP1 has been shown to be recruited to ...Missing: partners Src
  68. [68]
    Measuring Composition of CD95 Death-Inducing Signaling ...
    Aug 2, 2021 · DISC comprises DR, the adaptor protein Fas-associated protein with death domain (FADD), procaspases-8/-10, and cellular FADD-like interleukin ( ...
  69. [69]
    Structure of the Fas/FADD complex: A conditional death domain ...
    Dec 4, 2009 · This model portrays how sole death domains are able to mediate signaling upon receptor clustering in the complete absence of enzyme activity.Missing: Cryo- EM
  70. [70]
    Regulation of CD95/Fas signaling at the DISC - PubMed Central - NIH
    Nov 11, 2011 · This review is focused on the role in the CD95-mediated signaling of the death effector domain-containing proteins procaspase-8 and c-FLIP.
  71. [71]
    Daxx, a Novel Fas-Binding Protein That Activates JNK and Apoptosis
    We have identified a novel signaling protein, termed Daxx, that binds specifically to the Fas death domain. Overexpression of Daxx enhances Fas-mediated ...
  72. [72]
    The Death Receptor Pathway of Apoptosis - PMC - NIH
    The DISC forms within seconds of CD95 ... 2010. The Fas-FADD death domain complex structure reveals the basis of DISC assembly and disease mutations.
  73. [73]
    Ubiquitination and degradation of the FADD adaptor protein ... - Nature
    Jul 31, 2012 · Here we show that FADD is regulated by Makorin Ring Finger Protein 1 (MKRN1) E3 ligase-mediated ubiquitination and proteasomal degradation.Depletion Of Mkrn1 Induces... · Fadd Ubiquitination And... · Mkrn1 Depletion Facilitates...
  74. [74]
    Organization and Dynamics of Fas Transmembrane Domain in Raft ...
    Fas is recruited and clusters in lipid rafts after ligand-binding, a process that is vital for efficient DISC formation and apoptosis triggering (6,7,11). This ...
  75. [75]
    Cdk1/Cyclin B1 Controls Fas-Mediated Apoptosis by Regulating ...
    In this study, we demonstrate that procaspase-8 is phosphorylated in mitotic cells by Cdk1/cyclin B1 on Ser-387, which is located at the N terminus of the ...
  76. [76]
    Fas ligand mediates activation-induced cell death in human T ...
    We demonstrate that these two pathways of apoptosis are causally related. Stimulation of previously activated T cells resulted in the expression of Fas-L mRNA ...Missing: seminal paper
  77. [77]
  78. [78]
  79. [79]
    Non-apoptotic FAS signaling controls mTOR activation ... - PubMed
    Jan 12, 2024 · Non-apoptotic FAS signaling controls mTOR activation and extrafollicular maturation in human B cells. Sci Immunol. 2024 Jan 12;9(91):eadj5948.Missing: receptor | Show results with:receptor
  80. [80]
    Delayed Fas-mediated hepatocyte apoptosis during liver ... - PubMed
    Fas-mediated apoptosis is one of the major death processes of hepatocytes in liver diseases. Although compensatory regeneration occurs during liver injury, ...Missing: FasL | Show results with:FasL
  81. [81]
    Shedding of Fas ectodomain that affects apoptosis of hepatocytes ...
    Fas ectodomain shedding might be an important mechanism in controlling hepatocyte apoptosis during liver regeneration.
  82. [82]
    [PDF] Role of death receptors in the regulation of hepatocyte proliferation ...
    Oct 29, 2015 · ABSTRACT. The balance between hepatocyte proliferation and apoptosis is critical for liver homeostasis during liver regeneration.<|separator|>
  83. [83]
    3. Fas‐induced apoptosis in brian primary cultures - PubMed Central
    These results suggest that Fas is involved in neuronal apoptosis in the developing human brain. Keywords: Fas (APO‐1/CD95), apoptosis, human development, brain ...
  84. [84]
    Fas/Apo [Apoptosis]-1 and Associated Proteins in the Differentiating ...
    Mar 1, 1999 · ... Fas induces cell death by activation of caspase mechanisms (Nagata, 1997). Only ICEIII attenuated anti-Fas antibody-induced cell loss ...
  85. [85]
    Dynamic Fas signaling network regulates neural stem cell ... - Science
    Apr 22, 2020 · Our results demonstrate a novel Fas signaling network in the dentate gyrus and illuminate its consequences for adult neurogenesis and memory enhancement.
  86. [86]
    Non-apoptotic signaling pathways activated by soluble Fas ligand in ...
    Dec 14, 2001 · Non-apoptotic signaling pathways activated by soluble Fas ligand in serum-starved human fibroblasts. Mitogen-activated protein kinases and ...Missing: promotes migration wound healing
  87. [87]
    The cleaved FAS ligand activates the Na+/H+ exchanger NHE1 ...
    Jun 15, 2016 · We could observe that the cleaved ligand enhanced migration both in Boyden and Wound Healing experiments and that this effect was blocked by 10 ...
  88. [88]
    003234 - Fas- Strain Details - The Jackson Laboratory
    Nov 21, 2002 · Lymphadenopathy and splenomegaly in FAS deficient mice are evident at 8 weeks of age. The lymphnodes and spleen become progressively larger ...Missing: tissue | Show results with:tissue
  89. [89]
    Gene ResultFas Fas cell surface death receptor [ (house mouse)]
    Sep 24, 2025 · Data show that mutation of an inhibitory wedge motif in CD45 and deletion of Fas cooperate in mice, causing early lethality, autoantibody ...Missing: mammals | Show results with:mammals
  90. [90]
    Characterizing the regulatory Fas (CD95) epitope critical for agonist ...
    Oct 14, 2023 · Most biochemical studies have indicated higher affinity and binding priority for the inhibitory receptors over stimulatory receptors [50].
  91. [91]
    In vitro efficacy of Fas ligand gene therapy for the ... - PubMed - NIH
    Three human bladder cancer lines (T24, J82, and 5637) were treated with the conventional Fas agonist CH-11, a monoclonal antibody to the Fas receptor. Cells ...
  92. [92]
    Two Adjacent Trimeric Fas Ligands Are Required for Fas Signaling ...
    The proapoptotic TNF family member Fas ligand (FasL) signals cell death by engagement of its cognate receptor, Fas. The intracellular portion of Fas ...
  93. [93]
    Development of improved soluble inhibitors of FasL and CD40L ...
    In functional assays, Fas:COMP was at least 20-fold more active than Fas:Fc at inhibiting the action of sFasL, and CD40:COMP could block CD40L-mediated ...
  94. [94]
    Dissecting DISC regulation via pharmacological targeting of ...
    Jan 20, 2020 · In particular, the model has demonstrated that boosting caspase-8 activity by the small molecule at the early time points after DISC assembly is ...
  95. [95]
    Cyclosporin A abolishes CD28-mediated resistance to CD95 ...
    Dec 1, 2006 · In this study, we show that the immunosuppressive drug cyclosporin A (CsA) fully reverses resistance to CD95-mediated cell death after TCR/CD28 ...
  96. [96]
    Glucocorticoids in T cell apoptosis and function - PMC - NIH
    Glucocorticoids (GCs) are a class of steroid hormones which regulate a variety of essential biological functions. The profound anti-inflammatory and ...
  97. [97]
    A series of Fas receptor agonist antibodies that demonstrate ... - NIH
    Jan 20, 2012 · In this study, we identified a fully human anti-Fas antibody that could efficiently trigger apoptosis and therefore function as a potent agonist.
  98. [98]
    The abscopal effect of anti-CD95 and radiotherapy in melanoma
    May 16, 2023 · The anti-CD95 treatment plus radiation (combination treatment) reduced growth rates of both primary and secondary tumors relative to the control or radiation ...The Abscopal Effect Of... · 3.1 Anti-Cd95 Antibodies... · 3.2 Targeting Cd95 Enhances...
  99. [99]
    A Recombinant Bispecific CD20×CD95 Antibody With Superior ...
    Dec 15, 2015 · The bispecific antibody specifically triggers the CD95 death receptor on malignant, as well as activated, normal B-cells.
  100. [100]
    Potentiating CAR-T bystander killing by enhanced Fas/FasL ... - NIH
    Sep 24, 2025 · In this study, we modeled extreme Ag heterogeneity (>20%) to define how Fas/FasL-dependent bystander killing contributes to tumor clearance.Missing: emerging receptor fibrosis
  101. [101]
    Lipid Nanoparticle Fas Gene Therapy Suppresses Autoimmune ...
    Sep 15, 2025 · In the ALPS mice, loss of Fas function leads to decreased hematopoietic cell spontaneous apoptosis and mass accumulation of lymphocytes. To ...
  102. [102]
    IgG-Based Bispecific Anti-CD95 Antibodies for the Treatment of B ...
    Aug 16, 2022 · IgG-based bispecific anti-CD95 antibodies for the treatment of B cell-derived malignancies and autoimmune diseases.
  103. [103]
    A non-apoptotic caspase-8–meteorin pathway in hepatocytes ...
    Sep 26, 2025 · ... Fas-mediated caspase-3 and caspase-8 cleavage, did not impede ... A non-apoptotic caspase-8–meteorin pathway in hepatocytes promotes MASH fibrosis ...
  104. [104]
    Abrogation of Fas-induced fulminant hepatic failure in mice by ...
    Mar 27, 1998 · Administration of agonistic anti-Fas antibody to mice rapidly led to massive liver apoptosis and fulminant hepatic failure. In contrast ...Missing: challenges biomarkers stoichiometry