Fact-checked by Grok 2 weeks ago

Decay-accelerating factor

Decay-accelerating factor (DAF), also known as CD55, is a (GPI)-anchored membrane that functions as a primary regulator of the by accelerating the decay of and convertases in both the classical and alternative pathways, thereby preventing the formation of the membrane attack complex and protecting host cells from autologous complement-mediated lysis. Discovered in 1969 by E.M. Hoffmann, who isolated it from erythrocytes as a heat-stable factor inhibiting complement activity, DAF is encoded by the CD55 gene on chromosome 1q32 and exists in both membrane-bound and soluble forms. Structurally, DAF consists of four complement control protein (CCP) modules, also known as short consensus repeats (SCRs), each approximately 60 long, connected by short linkers and suspended above the by a serine//proline-rich region and a GPI anchor. The protein's molecular weight ranges from 50 to 100 kDa due to , extensive O- in the stalk region, and a single N-linked at 61 between CCPs 1 and 2, though this glycosylation is not essential for its regulatory function. CCPs 2 and 3 are primarily responsible for binding and dissociating the classical pathway (C4b2a), while CCPs 2, 3, and 4 mediate activity against the alternative pathway convertase (C3bBb); key residues such as the hydrophobic leucine-phenylalanine pair (L147F148) in CCP3 and the positively charged cluster (KKK125–127) between CCPs 2 and 3 are critical for these interactions. DAF is broadly expressed on the surface of most human cells, including erythrocytes, leukocytes, endothelial and epithelial cells, as well as in soluble forms in , , , , and , ensuring widespread protection against inadvertent complement activation. Beyond complement regulation, DAF modulates innate and adaptive immunity by suppressing T-cell and natural killer () cell activity through interactions with and serves as a for the adhesion CD97, influencing and signaling. It also acts as a receptor for pathogens, notably facilitating invasion by Plasmodium falciparum in , where CD55-null erythrocytes show resistance to infection. In disease contexts, DAF deficiency underlies conditions like (PNH), characterized by GPI-anchor defects leading to increased complement-mediated , and complement hyperactivation-associated protein-losing enteropathy (CHAPLE), resulting in gastrointestinal symptoms and due to unchecked complement activity; CHAPLE is treated with pozelimab (Veopoz), approved by the FDA in 2023 as the first specific therapy. Conversely, DAF is frequently overexpressed in various malignancies, including colorectal, , ovarian, and cancers, promoting tumor immune evasion and progression, which has positioned it as a potential therapeutic target for complement-enhancing anticancer strategies. Additionally, DAF contributes to autoimmune disorders such as and via CD55-CD97 interactions that exacerbate inflammation.

Molecular structure

Protein domains

The decay-accelerating factor (), also known as CD55, is a composed of 381 in its precursor form, including an N-terminal of 34 that directs translocation to the and a C-terminal (GPI) signal sequence of approximately 28 that facilitates membrane anchoring. The mature extracellular domain spans 319 , comprising four complement control protein (CCP) domains, also termed short consensus repeats (SCR), followed by a serine/threonine-rich stalk region rich in sites. Each of the four CCP domains (CCP1–CCP4) consists of approximately 60 and exhibits a conserved modular architecture typical of the regulators of complement activation () family. These domains feature two intramolecular bonds formed by four conserved residues, which stabilize a compact β-barrel-like fold comprising up to eight antiparallel β-strands arranged in a hydrophobic core, often anchored by a conserved residue. The β-sheet structure creates concave and convex faces, with the concave face implicated in interactions, while flexible loops and turns account for about half of each domain's residues, contributing to functional adaptability. The CCP domains play distinct yet cooperative roles in DAF's complement regulatory function, primarily by binding to C3b and C4b fragments to accelerate the decay of and convertases. CCP2 and CCP3 form the core for C3b and C4b, with CCP2 containing a positively charged region (residues 125–127) essential for pathway regulation and CCP3 featuring a hydrophobic pocket (residues 147–148) critical for decay acceleration in both classical and pathways. CCP1 primarily contributes to structural stabilization and positioning of the , while CCP4 enhances activity specifically in the pathway by supporting the of Bb from C3bBb convertases, requiring all three CCP2–CCP4 for full efficacy. Structural studies, including (NMR) spectroscopy of the CCP2–CCP3 fragment and of DAF in complex with 7, reveal an extended, rod-like arrangement of the domains with significant inter-domain flexibility at linker regions, allowing hinge-like adjustments (tilt angles of 35–39°) to accommodate binding without rigid inter-module contacts. This flexibility, evidenced by residual dipolar couplings in NMR data, enables the concave faces of adjacent CCPs to align optimally for multivalent interactions with convertase surfaces, underscoring the modular design's role in efficient complement inhibition.

Anchoring and modifications

The decay-accelerating factor (), also known as CD55, is anchored to the via a () anchor, which lacks a and allows flexible lateral movement within the . The anchor consists of a moiety linked through a conserved core—comprising a , three mannoses, and a phosphoethanolamine bridge—to the C-terminal serine residue of the mature protein following proteolytic cleavage of the C-terminal signal sequence. This ethanolamine-linked structure integrates into the outer leaflet of the plasma membrane, facilitating its role in surface protection without intracellular signaling domains. A prominent feature of DAF is extensive in its serine/threonine-rich stalk region, spanning approximately amino acids 261 to 319, which contains 14–16 O-linked oligosaccharide chains primarily composed of GalNAc-based mucin-type structures. These modifications add approximately 20 kDa to the protein's mass and serve a protective function by shielding the stalk from attack, thereby enhancing DAF stability on the surface. In contrast, N-linked is limited to a single site at 61 within the first complement control protein (CCP1) domain, featuring a complex biantennary that contributes to proper folding and solubility but accounts for only a minor portion of the total carbohydrate content. The overall post-translational modifications result in an apparent molecular weight of ~70 for membrane-bound DAF on , substantially higher than the predicted 41 from its sequence due to the combined ~30 from glycans. Variations in GPI anchor density across cell types or under pathological conditions can modulate DAF's surface mobility, with higher densities promoting nanoclustering in lipid rafts and improved diffusion to complement activation sites, while lower densities may reduce functional efficiency.

Genetics and expression

CD55 gene

The CD55 gene, which encodes the decay-accelerating factor (DAF) protein, is located on the long arm of chromosome 1 at the q32.2 cytogenetic band. It spans approximately 40 kb of genomic DNA and comprises 14 exons, as determined by detailed genomic analysis. This organization places CD55 within a chromosomal locus that also harbors other genes encoding complement regulatory proteins, facilitating coordinated regulation of complement pathways. The exon-intron structure of CD55 reflects the modular architecture of the DAF protein. Exon 1 encodes the N-terminal responsible for directing the nascent polypeptide to the secretory pathway. Exons 2 through 5 each encode one of the four complement control protein (CCP) domains (CCP1–CCP4), which are short consensus repeats critical for ligand binding. Exon 6 encodes the serine/threonine-rich stalk region that provides flexibility to the extracellular domain. Finally, exons 7–9 encode the (GPI) anchor attachment signal, enabling membrane anchoring of the mature protein, while in later exons can generate soluble isoforms lacking the GPI moiety. This exon arrangement supports diverse transcript variants, with at least 10 protein-coding isoforms identified. Polymorphisms in the CD55 gene primarily affect the blood group system (), a collection of 16 antigens expressed on erythrocytes and other cells via . These antigens arise from single polymorphisms (SNPs), predominantly in the exons encoding CCP domains, altering recognition without disrupting overall protein function in most cases. For instance, the rare Inab phenotype, characterized by complete absence of all Cromer antigens and reduced DAF expression, results from homozygous or frameshift , such as the W53X in 2 (c.159G>A), leading to premature termination and negligible protein production. Other Cromer variants, like (a–), stem from missense such as S165L in 5, which impair antigen expression but retain partial DAF activity. These genetic variants are recessive and have been mapped through family studies and sequencing of affected individuals. Transcriptional regulation of CD55 is responsive to inflammatory signals, particularly cytokines involved in immune . The gene's promoter region contains elements that mediate upregulation in response to interferon-gamma (IFN-γ), a key proinflammatory produced during and . In cell types such as colonic epithelial cells and certain tumor lines, IFN-γ treatment increases CD55 mRNA and surface protein levels, enhancing complement protection under stress conditions. This -inducible expression helps modulate local immune responses by bolstering membrane complement regulators on host cells. Additional cytokines like tumor necrosis factor-alpha (TNF-α) can synergize or antagonize this effect depending on the cellular context, underscoring CD55's role in adaptive immune .00290-1)

Tissue distribution

Decay-accelerating factor (), also known as CD55, exhibits ubiquitous expression across human cell types, serving as a key membrane-bound regulator of complement activation on the cell surface. It is prominently present on hematopoietic cells, including erythrocytes, leukocytes such as monocytes and granulocytes, and other blood cells, where it protects against autologous complement-mediated lysis. In non-hematopoietic lineages, is widely distributed on cells routinely exposed to complement, notably endothelial and epithelial cells lining vascular and mucosal surfaces. This broad localization underscores its role in safeguarding diverse tissues from inadvertent immune damage. Particularly high levels of DAF expression are observed on vascular , where it contributes to the integrity of walls amid constant complement exposure, and in the , especially at the feto-maternal on extravillous trophoblasts throughout . In contrast, neuronal cells display relatively low constitutive DAF expression, though it can be induced in response to inflammatory or stress signals, such as hypoxia-reoxygenation. During erythroid development, DAF expression on erythrocytes diminishes as cells mature, reflecting a shift in profiles post-differentiation. DAF expression is dynamically regulated, with upregulation occurring in response to inflammatory stimuli via transcription factors like , as seen in cytokine-activated endothelial and immune cells. Additionally, a soluble isoform (sDAF) is secreted from certain tissues, including epithelial linings, and circulates in , , , , and , potentially extending complement inhibitory functions beyond cell surfaces.

Biological function

Complement regulation

The decay-accelerating factor (), also known as CD55, serves as a key membrane-bound regulator of the by inhibiting the activation of the classical and pathways at the level of and convertases. DAF binds directly to the C3b and C4b components of these convertases, accelerating their dissociation into inactive forms: specifically, it promotes the release of C2a from the classical pathway convertase C4b2a and Bb from the pathway convertase C3bBb. This decay-accelerating activity occurs at a rate approximately 10- to 20-fold faster than spontaneous dissociation, thereby limiting the amplification of complement activation and preventing excessive deposition of C3b on host cells. By disrupting convertase stability, DAF reduces the generation of downstream effectors such as anaphylatoxins (C3a and C5a) and the membrane attack complex (MAC), protecting self-tissues from inadvertent . In addition to its decay-accelerating function, DAF acts as a cofactor for the factor I, facilitating the proteolytic cleavage of surface-bound C3b to its inactivated form iC3b and C4b to C4d. This cofactor activity further dampens complement amplification by inactivating the opsonin C3b, which would otherwise promote and further convertase assembly, while iC3b retains limited opsonic potential but cannot sustain the . Unlike some regulators, DAF's dual mechanisms—decay acceleration and cofactor function—target both pathways without directly inhibiting MAC formation, making it essential for early-stage control of complement on cell surfaces. DAF exhibits species-specific activity in complement regulation, with human DAF being ineffective at inhibiting porcine complement components, a limitation that has implications for where porcine organs are at risk from human complement attack. Native porcine regulators fail to adequately control human complement, necessitating the expression of human DAF in transgenic pigs to protect grafts from hyperacute rejection. DAF functions in synergy with other complement regulators, such as , which specifically inhibits assembly by binding C9 and preventing pore formation, and membrane cofactor protein (MCP or ), which provides cofactor activity but lacks decay-accelerating capability. This coordinated action ensures comprehensive inhibition across the complement cascade: DAF and MCP at the convertase stage, complemented by at the terminal stage.

Immune modulation

Beyond its primary function in complement inhibition, decay-accelerating factor (, also known as CD55) exerts complement-independent effects on immune s, influencing innate and adaptive responses through surface interactions and signaling pathways. serves as a ligand for the CD97, which is expressed on leukocytes including monocytes, granulocytes, and activated T and B s. This interaction functions as a costimulatory signal, enhancing T activation by promoting proliferation of + T s when co-engaged with the (CD3), as demonstrated in peripheral blood studies where CD97-mediated stimulation of CD55 increased expression of activation markers such as and CD25. Furthermore, the CD55-CD97 binding facilitates leukocyte and , supporting T recruitment to inflammatory sites without interfering with 's complement regulatory activity. In innate immune modulation, DAF contributes to polarization toward an anti-inflammatory phenotype through signaling cascades independent of complement. Upregulation of DAF represses (TLR) signaling and autocrine complement receptor (C3aR1/C5aR1) activation, thereby enabling activity, cyclic AMP () production, and (PKA)-mediated phosphorylation of CREB, which in turn supports Krüppel-like factor 4 ()-driven expression of markers like arginase-1 (Arg-1) and resistin-like alpha (Retnla). This mechanism suppresses proinflammatory responses, including reduced expression of chemoattractant protein-1 (MCP-1) and inducible (iNOS), as evidenced in DAF-deficient s where polarization is impaired. Such polarization fosters a tolerogenic , linking DAF to resolution of inflammation. DAF also modulates adaptive signaling, particularly by regulating production in T cells. In murine models of , DAF deficiency results in hypersecretion of interleukin-2 (IL-2) by restimulated T cells, indicating that DAF normally suppresses IL-2 output to temper T cell responses; this effect is complement-dependent, as it is reversed by or blockade. Elevated IL-2 in DAF-null contexts persists in memory T cells, highlighting DAF's role in fine-tuning T cell immunity during recall responses. In the context of tumorigenesis, overexpression on cancer cells promotes immune evasion by diminishing . By accelerating decay of convertases, limits C3b/iC3b deposition on tumor surfaces, thereby reducing recognition and engulfment by macrophages and other , as observed in colorectal, breast, and ovarian cancers where high correlates with poor prognosis and resistance to immune clearance. This protective mechanism extends to inhibition of activity, further shielding tumors from innate surveillance.

Disease associations

Paroxysmal nocturnal hemoglobinuria

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematopoietic stem cell disorder caused by a somatic mutation in the PIGA gene, which encodes a key enzyme in the biosynthesis of glycosylphosphatidylinositol (GPI) anchors. This mutation, occurring in hematopoietic stem cells, results in the absence or severe deficiency of GPI-anchored proteins on the surface of blood cells, including decay-accelerating factor (DAF, or CD55) and membrane inhibitor of reactive lysis (CD59). Without these complement regulators, affected red blood cells, granulocytes, and platelets become highly susceptible to complement-mediated lysis, leading to the hallmark intravascular hemolysis of PNH. The PIGA mutation is X-linked and typically affects a single stem cell clone, which expands over time, with the proportion of PNH-type cells varying among patients. The clinical manifestations of PNH arise primarily from uncontrolled complement activation due to and deficiency. Intravascular is chronic, with paroxysmal exacerbations that were historically termed "nocturnal" because concentrated urine in the morning made more noticeable, though the itself is continuous rather than sleep-specific. This leads to symptoms such as , dyspnea, , and dark urine from . , a major cause of morbidity and mortality, occurs in up to 40% of patients, often in unusual sites like , due to platelet activation and free hemoglobin's procoagulant effects. failure, present in about 30-50% of cases, contributes to cytopenias and may overlap with or myelodysplastic syndromes. PNH has an estimated incidence of 1-2 cases per million individuals annually. Diagnosis of PNH relies on flow cytometry to detect the absence of CD55 and CD59 on peripheral blood cells, particularly granulocytes and monocytes, which provides high sensitivity and specificity for identifying the PNH clone size. Red blood cells are classified into type I (normal GPI expression), type II (partial deficiency), and type III (complete absence), with type III cells predominant in hemolytic PNH. Historical tests like the Ham acid hemolysis test, which demonstrated increased red cell lysis in acidic conditions, have been largely replaced by flow cytometry due to their lower sensitivity and technical demands. Treatment targets complement inhibition and, in select cases, addresses the underlying clonal disorder. , a against complement component , blocks the terminal complement pathway, dramatically reducing intravascular , transfusion requirements, and risk in most patients, as shown in pivotal trials. Long-acting C5 inhibitors like offer similar benefits with less frequent dosing. More recently, as of 2025, proximal complement inhibitors such as (approved 2023) and danicopan (approved 2024), which target factor B or factor D to inhibit earlier in the pathway, and crovalimab (approved 2024), a recycling inhibitor, have been approved, providing options for both intravascular and extravascular with oral or . remains a foundational , particularly during . Allogeneic is the only curative option, eradicating the PNH clone and restoring normal hematopoiesis, particularly recommended for young patients with significant failure or refractory disease. Supportive care, including anticoagulation for and iron supplementation for hemoglobinuria-induced losses, complements these approaches.

Complement hyperactivation-associated protein-losing enteropathy (CHAPLE)

Complement hyperactivation-associated protein-losing enteropathy (CHAPLE) is a rare autosomal recessive disorder caused by biallelic loss-of-function mutations in the CD55 gene, leading to deficiency of membrane-bound and resultant hyperactivation of the . First described in 2017, CHAPLE typically presents in early childhood with gastrointestinal symptoms including chronic diarrhea, abdominal pain, and due to intestinal from endothelial damage by uncontrolled complement. Additional features include recurrent infections, angiopathic (e.g., in cerebral or renal vessels), and from , with an estimated prevalence of less than 1 in 1,000,000. Diagnosis involves confirming CD55 mutations, showing absent on blood cells, and evidence of complement hyperactivation (e.g., low levels). focuses on complement inhibition; pozelimab (Veopoz), a , was approved by the FDA in 2023 as the first specific therapy, administered subcutaneously weekly after an initial intravenous dose, significantly reducing protein loss and risk in clinical trials. Supportive measures include nutritional support, anticoagulation, and infection prophylaxis.

Infectious diseases

Decay-accelerating factor (, also known as CD55) serves as a cellular receptor for several picornaviruses, facilitating their attachment and entry into host cells. Specifically, DAF binds coxsackie B viruses and echoviruses through its complement control protein domains CCP3 and CCP4, which interact with viral proteins to promote . This interaction is critical for infection, as mutations in these domains abolish viral binding. Similarly, certain serotypes of 70 utilize DAF as a primary receptor on ocular and respiratory epithelial cells, enabling and respiratory infections. In , DAF acts as a receptor for merozoites, facilitating their invasion of human erythrocytes; erythrocytes lacking CD55 (e.g., in rare blood group phenotypes) show resistance to infection, highlighting DAF's role in parasite . In the context of human immunodeficiency virus type 1 (HIV-1), DAF contributes to viral evasion of complement-mediated destruction. HIV-1 particles incorporate DAF from infected host cells into their during , where it functions to destabilize and convertases, thereby inhibiting complement opsonization and neutralization in serum. This incorporation enhances viral survival in the bloodstream, as demonstrated by experiments showing reduced complement sensitivity in DAF-associated virions compared to those lacking it. Bacterial pathogens also exploit DAF to modulate host immune responses. interacts with DAF to dampen complement activation, thereby resisting by neutrophils and macrophages. In infection models, DAF provides protective effects for the host against certain streptococcal strains by regulating excessive , although its absence can paradoxically enhance granulocyte-mediated clearance in some cases. Experimental studies using DAF-deficient mice highlight its role in . In models of B3 infection, DAF mice exhibit altered susceptibility to , with reduced severity of cardiac compared to wild-type mice, underscoring DAF's contribution to viral and immune cell activation in the heart.

Clinical and research applications

Therapeutic uses

Recombinant soluble forms of decay-accelerating factor (sDAF) have been investigated as therapeutics to systemically inhibit complement activation and mitigate in complement-related disorders. Preclinical studies demonstrated that sDAF effectively suppresses immune complex-mediated in models resembling by accelerating the decay of and convertases. In , expression of in transgenic pigs has been a key strategy to counteract hyperacute rejection by protecting porcine endothelial cells from complement . These genetically modified pigs, incorporating alongside other regulators like and , have extended graft survival in preclinical nonhuman primate models. As of 2025, clinical trials are underway evaluating DAF-expressing gene-edited pig kidneys and hearts for transplantation, with initial procedures demonstrating feasibility and reduced early rejection in end-stage failure patients. Cancer therapies targeting DAF leverage its overexpression on tumor cells, which promotes immune evasion by inhibiting complement-mediated lysis. Anti-CD55 monoclonal antibodies enhance antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against DAF-expressing malignancies, such as colorectal tumors. For instance, a novel anti-CD55 antibody activated complement to suppress proliferation, invasion, and migration in colorectal cancer cells, with combination therapy alongside 5-fluorouracil amplifying antitumor effects. The 791Tgp72 antigen, identified as a glycosylated form of CD55, serves as a vaccine target in colorectal, gastric, and ovarian cancers, eliciting T-cell responses and supporting imaging and immunotherapy approaches. In (PNH), where deficiency exacerbates complement-driven , -mimicking strategies complement inhibitors by targeting upstream convertase activity to prevent opsonization and residual extravascular . Proximal inhibitors, such as factor D blockers like danicopan, are combined with inhibitors (e.g., or ) to restore regulation akin to function, improving levels and reducing transfusion needs in patients with incomplete responses to monotherapy. This adjunctive approach addresses the multifaceted complement dysregulation in PNH, enhancing overall disease control.

Pathogen interactions

Research on decay-accelerating factor (DAF, also known as CD55) as a target for interventions has highlighted its role as a cellular receptor exploited by various , enabling strategies to disrupt -host interactions and enhance immune clearance. In antiviral approaches, soluble DAF-Fc fusion proteins have demonstrated efficacy in blocking binding and infection. Specifically, DAF-Fc inhibits B3 (CVB3) infection by competing with cell-surface DAF for viral attachment, reducing plaque formation and across multiple strains. In vivo, administration of DAF-Fc in models of CVB3-induced attenuates in the heart, decreases myocardial , and improves survival rates when given early post-infection, suggesting its potential as a therapeutic trap for enteroviral diseases. These findings have prompted exploration of DAF-targeted receptor traps for development against coxsackieviruses, where soluble forms could neutralize circulating and elicit protective immunity without full . In research, incorporated into virions during budding from infected cells shields the virus from complement-mediated , contributing to immune evasion. Soluble constructs have been investigated to counteract this protection; studies show that -Fc fusion proteins bind to HIV-1 particles, displacing membrane-anchored and restoring susceptibility to antibody-dependent complement-mediated , thereby enhancing viral under complement-sufficient conditions. This approach leverages 's dual role in attachment and complement regulation, potentially amplifying the efficacy of neutralizing antibodies in clearing HIV-infected cells. For bacterial pathogens, DAF serves as an adherence receptor for certain strains, and its diminishes in relevant models. Analogous mechanisms apply to like diffusely adhering (DAEC), which use Afa/Dr adhesins to bind DAF on epithelial cells, facilitating colonization and invasion. In vitro of DAF with monoclonal antibodies significantly reduces DAEC adherence to host cells, inhibiting formation and bacterial . Emerging investigations into DAF's involvement in reveal its upregulation in severe cases, correlating with dysregulated complement activation and poor outcomes. In patients with severe infection, CD55 expression is elevated on monocytes, + T cells, and + T cells, suppressing type I responses and promoting excessive via unchecked complement deposition. hijacks host CD55 for incorporation into virions, impairing antibody-dependent complement-mediated and exacerbating endothelial damage and . Therapeutic targeting of upregulated DAF, such as through monoclonal antibodies or soluble inhibitors, restores complement sensitivity , suggesting potential interventions to mitigate in critical by enhancing viral clearance and reducing hyperinflammation.

References

  1. [1]
    Beyond the Role of CD55 as a Complement Component - PMC
    Feb 20, 2018 · Decay-accelerating factor (DAF or CD55) is a known inhibitor of the complement system and has recently attracted substantial attention due to ...<|control11|><|separator|>
  2. [2]
    Structure/function studies of human decay-accelerating factor - PMC
    The decay-accelerating factor (DAF) contains four complement control protein repeats (CCPs) with a single N-linked glycan positioned between CCPs 1 and 2.
  3. [3]
    Decay-Accelerating Factor - an overview | ScienceDirect Topics
    Decay-Accelerating Factor ... The first identification of membrane factors that could regulate complement activation came from Hoffmann and colleagues in 1969.
  4. [4]
    Structure of decay-accelerating factor bound to echovirus 7: A virus ...
    Its extracellular region contains four 60-aa short consensus repeat (SCR) domains, also known as complement control protein (CCP¶) domains, followed by a 97-aa ...
  5. [5]
    Structure-Function Analysis of Decay-Accelerating Factor
    The repeat three of DAF was shown to be important in Dr adhesin binding and complement regulation.
  6. [6]
    Solution structure of a functionally active fragment of decay ... - NIH
    We report here the solution structure and dynamics of DAF∼2,3 providing a characterized structure of a mammalian protein with decay-accelerating activity.Missing: peptide | Show results with:peptide
  7. [7]
    Regulators of complement activity mediate inhibitory mechanisms ...
    The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through ...
  8. [8]
    Decay-accelerating Factor (DAF), Complement Receptor 1 (CR1 ...
    Both of these membrane-bound proteins require three CCPs for decay acceleration activity, CCP 2–4 of DAF and CCP 1–3 of CR1, with the two respective regions ...
  9. [9]
    GPI-anchored protein organization and dynamics at the cell surface
    FRAP-based studies also showed that apically sorted GPIs are immobile just after reaching the cell surface, and their mobility increases as they become long- ...
  10. [10]
    Analysis of the signal for attachment of a glycophospholipid ...
    The COOH terminus of decay accelerating factor (DAF) contains a signal that directs attachment of a glycophospholipid (GPI) membrane anchor.
  11. [11]
    Effects of O-linked glycosylation on the cell surface expression and ...
    Oct 15, 1989 · Decay accelerating factor (DAF) is a glycophospholipid-anchored membrane glycoprotein that protects mammalian host cells from inadvertant ...Missing: GPI glycan Ser318
  12. [12]
    Entry - *125240 - CD55 ANTIGEN; CD55 - OMIM - (OMIM.ORG)
    Gene Structure​​ Osuka et al. (2006) determined that the CD55 gene contains 14 exons.
  13. [13]
    Gene ResultCD55 CD55 molecule (Cromer blood group) [ (human)]
    Aug 19, 2025 · CD55 is upregulated by cAMP/PKA/AKT and modulates human decidualization via Src and ERK pathway and decidualization-related genes. Erythrocyte ...Missing: chromosome exons
  14. [14]
  15. [15]
    Gene: CD55 (ENSG00000196352) - Summary - Homo_sapiens
    This gene has 34 transcripts (splice variants), 97 orthologues, 39 paralogues and is associated with 3 phenotypes. Transcripts. Show transcript table ...
  16. [16]
    Entry - #613793 - BLOOD GROUP, CROMER SYSTEM; CROM - OMIM
    The red blood cells (RBCs) of people with the Cromer-null phenotype, Inab, lack DAF but do not appear to show increased susceptibility to hemolysis. Antibodies ...
  17. [17]
  18. [18]
    Expression and regulation by interferon-gamma of the membrane ...
    Upregulation of protein expression was only observed in HT29 cells for CD55 and CD59 and was accompanied by a marked increase of the corresponding transcripts.Missing: Liu et 2005
  19. [19]
    Cytokine-mediated up-regulation of CD55 and CD59 protects ... - NIH
    Interferon-gamma reduced expression of CD59 and strongly antagonized the up-regulatory effects on CD59 mediated by the other cytokines. Complement attack on ...
  20. [20]
    Complement decay accelerating factor (DAF)/CD55 in cancer - PMC
    We will review the expression of CRPs in cancer, focussing on CD55 and highlight other roles of the CRPs and their involvement in leukocyte function.
  21. [21]
    Complement Regulators: Decay-accelerating Factor | Sino Biological
    Anchoring of DAF to phospholipids on the cell membrane occurs through a glycosylphosphatidylinositol (GPI) moiety. Soluble forms of decay-accelerating factor/ ...
  22. [22]
    The complement system in human pregnancy and preeclampsia
    EVTs express CD46, CD55, and CD59 in all three trimesters of normal pregnancy (411, 412). The placentae obtained from women with preeclampsia overexpress CD55 ...
  23. [23]
    Decay accelerating factor (CD55) protects neuronal cells from ...
    Apr 9, 2010 · Human NT2-N neurons constitutively express DAF which is down-regulated after severe hypoxia and subsequent reoxygenation with human serum [14].
  24. [24]
    Expression of the DAF (CD55) and CD59 antigens during normal ...
    Aug 6, 2025 · With erythroid development, i.e., after loss of CD45 and decrease of CD71, expression of both proteins decreased. With myeloid maturation, ...
  25. [25]
    The genetic and epigenetic regulation of CD55 and its pathway ...
    Our results indicated that CD55 is regulated by TFCP2, NF-κB, miR-27a-3p, and several immune-related genes, which in turn affects colon cancer.
  26. [26]
    Identification of the complement decay-accelerating factor (DAF) on ...
    Mar 1, 1987 · Additionally, we detected soluble DAF antigen in plasma, tears, saliva, and urine, as well as in synovial and cerebrospinal fluids. While ...Missing: sDAF | Show results with:sDAF
  27. [27]
    Decay-accelerating factor must bind both components of ... - PubMed
    Jan 1, 2007 · Decay-accelerating factor (DAF; CD55) inhibits the complement (C) cascade by dissociating the multimolecular C3 convertase enzymes central to amplification.Missing: system | Show results with:system
  28. [28]
    The Role of Membrane Bound Complement Regulatory Proteins in ...
    May 21, 2019 · CD55, CD46, and CD59 are known to exert control on all three pathways of complement activation. CD55, also known as DAF, accelerates the decay ...
  29. [29]
    Prevention of hyperacute rejection by human decay accelerating ...
    As a potential source of organs for xenotransplantation, pigs that are transgenic for human decay accelerating factor (DAF) have been bred in order to overcome ...
  30. [30]
    Xenotransplantation: where are we today? - PMC - NIH
    In view of the putative species incompatibility of porcine complement regulating proteins, pigs have been generated which express human complement regulators.
  31. [31]
    The Inhibitory Effect of CD46, CD55, and CD59 on Complement ...
    Apr 1, 2002 · Only the combination of anti-CD55 and anti-CD59 resulted in an increase in lysis, and the effect was more additive than synergistic. These ...
  32. [32]
  33. [33]
  34. [34]
  35. [35]
  36. [36]
    Paroxysmal Nocturnal Hemoglobinuria: A Complement ... - PMC - NIH
    SUMMARY. PNH is caused by a somatic mutation in PIGA that leads to a marked deficiency or absence of the complement regulatory proteins CD55 and CD59. The ...
  37. [37]
    Paroxysmal nocturnal hemoglobinuria: pathophysiology, natural ...
    PIG-A mutations: mutation occurring in the PIG-A gene may affect the synthesis or the function of the encoded protein, resulting in the impairment of the first ...
  38. [38]
    Paroxysmal Nocturnal Hemoglobinuria - StatPearls - NCBI Bookshelf
    Traditionally PNH was characterized by recurrent episodes of intravascular hemolysis, venous thrombosis, and cytopenias associated with bone marrow failure.
  39. [39]
    The Incidence and Prevalence of Paroxysmal Nocturnal ...
    Nov 16, 2006 · 76 PNH patients were diagnosed during this time period giving an incidence of 0.13/100,000/year. Based on incidence and survival rates, the ...
  40. [40]
    Diagnosis of Paroxysmal Nocturnal Hemoglobinuria - PMC - NIH
    Various tests like modified Ham's test, sucrose lysis test, gel card technique (GCT) and flow cytometry-based testing are available for making diagnosis [9–12].
  41. [41]
    The Complement Inhibitor Eculizumab in Paroxysmal Nocturnal ...
    Excessive or persistent intravascular hemolysis in patients with PNH causes anemia, hemoglobinuria, and complications related to the presence of plasma free ...
  42. [42]
    Paroxysmal nocturnal hemoglobinuria: current treatments and ...
    Currently, the only cure for paroxysmal nocturnal hemoglobinuria (PNH) is an allogeneic hematopoietic stem cell transplantation. Stem cell transplantation ...
  43. [43]
    Stem cell transplantation for paroxysmal nocturnal hemoglobinuria
    Eculizumab is highly effective in reducing intravascular hemolysis in paroxysmal nocturnal hemoglobinuria and markedly reduces the risk for thrombosis;– it does ...
  44. [44]
    Echoviruses and coxsackie B viruses that use human decay ...
    Echoviruses and coxsackie B viruses that use human decay-accelerating factor (DAF) as a receptor do not bind the rodent analogues of DAF ... CD55 Antigens ...
  45. [45]
    Human Diffusely Adhering <italic toggle='yes'>Escherichia coli ...
    the majority of CD55-binding enteroviruses require CCP3 and. CCP4 (47), although EV7 binding may require CCP2, CCP3, and CCP4 (11, 21). The specificity of ...
  46. [46]
    The HeLa cell receptor for enterovirus 70 is decay-accelerating ...
    Decay-accelerating factor (DAF, CD55) is a 70- to 75-kDa GPI-anchored membrane protein that is involved in the regulation of complement and has also been ...
  47. [47]
    Role of virion-associated glycosylphosphatidylinositol-linked ... - NIH
    These results show that CD55 and CD59 are incorporated into HIV-1 particles and function to protect virions from complement-mediated destruction, and they are ...
  48. [48]
    Acquisition of Complement Resistance through Incorporation of ...
    Human immunodeficiency virus (HIV) is known to develop resistance to human complement through the incorporation of DAF, CD46, and CD59 to the viral particles ( ...
  49. [49]
    A Novel Role for CD55 in Granulocyte Homeostasis and Anti ... - NIH
    Oct 3, 2011 · Cd55-/- mice have an increased resistance against Streptococcal pneumoniae infection. Figure 4 · Open in a new tab. Wild-type and Cd55 -/- mice ...
  50. [50]
    A Novel Role for CD55 in Granulocyte Homeostasis and Anti ...
    These data suggest that complement-independent interaction of CD55 with CD97 is functionally relevant and involved in granulocyte homeostasis and host defense.
  51. [51]
    Decay-accelerating factor (CD55) promotes CD1d expression and ...
    ... Decay-accelerating factor (CD55) promotes CD1d expression and Vgamma4+ T-cell activation in coxsackievirus B3-induced myocarditis. Viral Immunol. 2006 Summer ...
  52. [52]
    Autoimmunity in Coxsackievirus B3 induced myocarditis
    Studies using DAF knockout mice confirm that this receptor is important for myocarditis but not pancreatitis [153]. DAF expression is enhanced by estrogen ...
  53. [53]
    Coupling complement regulators to immunoglobulin domains ... - NIH
    ... soluble DAF was released from DAF-Ig using papain. The sDAF produced this way had identical activity to sDAF secreted from CHO cells (Fig. 3b). The ability ...
  54. [54]
    Human recombinant soluble decay accelerating factor inhibits ...
    mDAF is a more potent inhibitor of C-mediated hemolysis than either sDAF or seDAF, suggesting that incorporation into cell membranes greatly enhances the ...
  55. [55]
    Controlling the complement system in inflammation - ScienceDirect
    Attempts to efficiently inhibit complement include the application of endogenous soluble complement inhibitors (C1-inhibitor, recombinant soluble complement ...
  56. [56]
    2025: status of cardiac xenotransplantation including preclinical ...
    Apr 15, 2025 · Hearts from transgenic pigs constructed with CD59/DAF genomic clones demonstrate improved survival in primates. Xenotransplantation. (1999) ...
  57. [57]
  58. [58]
    The generation and evaluation of TKO/hCD55/hTM/hEPCR gene ...
    Jan 20, 2025 · This study investigates the generation of TKO/hCD55/hTM/hEPCR (6GE) pigs and evaluates their compatibility with human immune and coagulation systems.<|separator|>
  59. [59]
    A novel therapeutic anti‑CD55 monoclonal antibody inhibits the ...
    The CD55 antibody treatment activated complement and therefore suppressed the proliferation, invasion and migration of colorectal cancer cells.
  60. [60]
    A novel therapeutic anti‑CD55 monoclonal antibody inhibits the ...
    Sep 26, 2019 · Among complement regulatory proteins, the present study focused on decay-accelerating factor (CD55 or DAF or 791Tgp72), an inhibitor of ...Statistical Analysis · Results · Upregulation Of Cd55 During...<|control11|><|separator|>
  61. [61]
    Decay Accelerating Factor (CD55): A Target for Cancer Vaccines?1
    CD55 inhibits the complement cascade by inactivating C3 convertases and preventing C3b deposition on cell membranes. C3b is a powerful stimulus of phagocytosis ...
  62. [62]
  63. [63]
    Paroxysmal nocturnal hemoglobinuria: Where are we going
    Feb 16, 2023 · In this case, combination treatments, with proximal complement inhibitors and C5 inhibitors, may be ideal for the treatment of PNH.
  64. [64]
    Pharmacological Therapies in Paroxysmal Nocturnal ... - NIH
    Sep 23, 2025 · The majority of cases of PNH are caused by a mutation of the PIGA gene [6–8]. PIGA is required for the formation of the glycophosphatidyl (GPI) ...Missing: etiology | Show results with:etiology
  65. [65]
    Inhibition of Coxsackie B Virus Infection by Soluble Forms of Its ...
    The top row represents virus infection of cells in the absence of inhibitor, while a fivefold dilution of monomeric DAF (1.0 μM to 16 nM) or dimeric DAF-Fc (0.5 ...
  66. [66]
    Soluble Recombinant Coxsackievirus and Adenovirus Receptor ...
    We have shown elsewhere that early DAF-Fc treatment attenuates CVB3-induced myocarditis and virus replication. Methods: CAR was synthesized as a soluble IgG1-Fc ...
  67. [67]
    Coxsackievirus B3-Associated Myocardial Pathology and Viral Load ...
    DAF-Fc may be a novel therapeutic agent for active myocarditis and acute dilated cardiomyopathy if given early in the infectious period.
  68. [68]
    A novel approach to inhibit HIV-1 infection and enhance lysis of HIV ...
    This regulation is mediated by proteins such as cell surface-like membrane cofactor protein (MCP), decay accelerating factor (DAF) and protectin (CD59), and the ...
  69. [69]
    HIV and human complement: inefficient virolysis and effective ...
    In this review, we focus on the interaction of HIV with the human complement system, regarding the mechanisms responsible for the inefficient complement- ...
  70. [70]
    Binding of Escherichia coli adhesin AfaE to CD55 triggers ... - PNAS
    For blockade of bacterial adhesion, CD55-specific mAb BRIC216 or an Ig-isotypic control (10 μg/ml) was incubated with targets before bacterial infection. In ...
  71. [71]
    Pathogenesis of Afa/Dr Diffusely Adhering Escherichia coli - PMC
    In this review, the focus is on Afa/Dr DAEC strains that have been found to be associated with urinary tract infections and with enteric infection.<|control11|><|separator|>
  72. [72]
    Upregulation of CD55 complement regulator in distinct PBMC ...
    Oct 7, 2022 · The present study identifies a COVID-19 specific CD55 expression pattern in PBMC subpopulations that coincides with reduced interferon responses.Results · Pbmc Transcriptomic... · Cd55 Upregulation In Severe...
  73. [73]
    SARS-CoV-2 hijacks host CD55, CD59 and factor H to impair ...
    Oct 22, 2024 · These include the plasma membrane-resident protectin (CD59) and decay accelerating factor (DAF, CD55) as well as the fluid-phase regulator ...
  74. [74]
    Full article: SARS-CoV-2 hijacks host CD55, CD59 and factor H to ...
    Here, we demonstrate that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hijacks host cellular RCA members CD55 and CD59 and serum-derived Factor ...