FCAR Human

Fc Fragment Of IgA Receptor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cellular Distribution

Cell TypeExpressionNotes
NeutrophilsHigh (circulating)Downregulated in tissue macrophages
MonocytesModerateDownregulated during macrophage maturation
EosinophilsPresentRole in parasitic immunity
MacrophagesAbsent (intestine), Alveolar (a.2 variant)Tissue-specific isoforms

Regulation:

  • Primed by PI3K/p38/PKC pathways via cytokine signaling .

  • Shedding of polymeric IgA/FCAR complexes reported in circulation .

Pro-Inflammatory Signaling

PathwayMechanismOutcomes
ITAM ActivationLyn-dependent phosphorylation → Syk recruitment → PI3K/PLC-γ activationCytokine release (TNF-α, IL-6), Phagocytosis, NETosis
Sustained AggregationCrosslinking of FcαRI → Full ITAM phosphorylationADCC, ADCP, Antigen presentation

Anti-Inflammatory Signaling

PathwayMechanismOutcomes
ITAMi (Inhibitory ITAM)Partial FcRγ phosphorylation → SHP-1 recruitment → Inhibition of FcγR/FcεRSuppression of degranulation, Immune homeostasis
Monomeric IgA BindingTransient interaction → FcRγ-independent endocytosisRecycling to cell surface, Anti-inflammatory priming

Splice Variants and Functional Diversity

VariantDomain AlterationExpression SiteFunction
a.2Lacks 22 aa pre-TM domainAlveolar macrophagesUnique to lung tissue
a.3Lacks EC2 domainUndeterminedReduced IgA binding capacity

Note: Over 10 mRNA variants exist, but only a.2 and a.3 are validated at the protein level .

Species-Specific Differences and Research Models

SpeciesFCAR PresenceKey Features
HumanYesChromosome 19, dual IgA1/IgA2 isotypes, Mucosal/extracellular roles
MouseNoLacks homolog → Transgenic models developed (C57BL/6, BALB/c, SCID backgrounds)
Rat/HorseYes~55–58% aa identity with human FCAR

Transgenic Models:

  • Human FCAR inserted under endogenous promoter in mice to study IgA-mediated ADCC and ADCP .

  • Applications: Tumor immunology, Autoimmune disease modeling, Vaccine development .

Recombinant FCAR Proteins

SourceCharacteristicsApplications
Sf9 Baculovirus215 aa, 24.5 kDa, His-tag, Glycosylated (28–40 kDa on SDS-PAGE)Binding assays, Structural studies
Mammalian CellsFull-length (Gln22-Asn227), 6-His tag, K<sub>D</sub> <10 nMELISA, Flow cytometry, Receptor-ligand studies

Experimental Notes:

  • Carrier-free versions avoid BSA interference in biochemical assays .

  • Shedding and recycling kinetics studied via labeled IgA/FCAR complexes .

Disease Associations

ConditionRole of FCAREvidence
Inflammatory Bowel DiseaseModulation of mucosal IgA responsesAltered expression in intestinal macrophages
Autoimmune DiseasesAnti-inflammatory signaling via ITAMiSHP-1 recruitment limits excessive inflammation
CancerADCC by neutrophils against IgA-opsonized tumorsCytokine release (TNF-α, IL-1β) enhances immune infiltration

Product Specs

Introduction

The Fc Fragment Of IgA Receptor, also known as FCAR, is a member of the multichain immune recognition receptor family. IgA, the antibody that binds to FCAR, is the most abundant immunoglobulin in mucosal areas, though it is the second most common antibody isotype in serum. FCAR plays a role in both pro-inflammatory and anti-inflammatory responses depending on the state of the IgA that is bound to it. FCAR is also a key Fc receptor for the neutrophil-mediated killing of tumor cells. When neutrophils expressing FCAR interact with tumor cells that have been opsonized with IgA, the neutrophils exhibit antibody-dependent cell-mediated cytotoxicity and release the cytokines TNF-α and IL-1β, which lead to increased neutrophil migration to the tumor site.

Description

Produced in Sf9 Baculovirus cells, FCAR is a single, glycosylated polypeptide chain with a molecular weight of 24.5 kDa. It consists of 215 amino acids (22-227a.a.). On SDS-PAGE, the molecular size will appear to be approximately 28-40 kDa. FCAR is expressed with a 9 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.

Physical Appearance
Sterile Filtered colorless solution.
Formulation

The FCAR protein solution (0.25 mg/mL) is supplied in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.

Stability

For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.

Purity

Purity is determined to be greater than 85.0% by SDS-PAGE analysis.

Synonyms

Fc Fragment Of IgA Receptor, Fc Fragment Of IgA, Receptor For, CD89, Immunoglobulin Alpha Fc Receptor, Fc Alpha Receptor, FCAR Variant 14, IgA Fc Receptor, CD89 Antigen, CTB-61M7.2, FcalphaRI, Immunoglobulin alpha Fc receptor, IgA Fc receptor.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPQEGDFPM PFISAKSSPV IPLDGSVKIQ CQAIREAYLT QLMIIKNSTY REIGRRLKFW NETDPEFVID HMDANKAGRY QCQYRIGHYR FRYSDTLELV VTGLYGKPFL SADRGLVLMP GENISLTCSS AHIPFDRFSL AKEGELSLPQ HQSGEHPANF SLGPVDLNVS GIYRCYGWYN RSPYLWSFPS NALELVVTDS IHQDYTTQNH HHHHH.

Q&A

What is the structural organization of human FCAR/CD89?

Human FCAR/CD89 is a type I transmembrane glycoprotein belonging to the multichain immune recognition receptor (MIRR) family. The protein contains a 21 amino acid signal sequence followed by distinct domains: an extracellular domain (ECD) of 206 amino acids, a transmembrane (TM) domain of 19 amino acids, and a cytoplasmic domain of 41 amino acids . The ECD consists of two C2-type immunoglobulin-like domains (EC1 and EC2) that are oriented at right angles to each other, with EC1 being primarily responsible for IgA binding . The transmembrane region contains an arginine residue at position 230 (Arg230) that is critical for association with the ITAM-containing FcR gamma signaling subunit, which contains a complementary transmembrane aspartic acid residue . This structural organization is essential for understanding how FCAR mediates its biological functions and for designing experiments to investigate receptor-ligand interactions and downstream signaling events.

How can I determine FCAR/CD89 expression in different cell populations?

FCAR/CD89 expression can be reliably determined through several complementary methodological approaches. At the protein level, flow cytometry represents the gold standard for detecting FCAR/CD89 on cell surfaces, allowing researchers to simultaneously identify specific cell populations and quantify receptor expression levels . For this application, validated antibodies such as sheep anti-human FCAR/CD89 antigen affinity-purified polyclonal antibody or Alexa Fluor 405-conjugated antibodies can be utilized . Western blotting provides an alternative method for detecting total FCAR protein in cell lysates, allowing assessment of different glycosylation forms that typically appear as bands between 50-100 kDa . At the transcript level, quantitative PCR using specific primers and probes, such as the PrimePCR probe assay, enables precise measurement of FCAR gene expression . For comprehensive tissue distribution analysis, immunohistochemistry can be employed to visualize FCAR expression in tissue sections. It's important to note that FCAR expression varies significantly among myeloid lineages, with strong expression on circulating neutrophils, eosinophils, and monocytes, but downregulation occurs as monocytes differentiate into tissue macrophages .

What are the key splice variants of FCAR/CD89 and how do they differ functionally?

Multiple alternatively spliced transcript variants of FCAR/CD89 have been reported in the literature, though only two have been definitively identified at the protein level . The a.2 variant, which lacks 22 amino acids immediately preceding the transmembrane domain, exhibits a highly specific expression pattern, being exclusively found in alveolar macrophages . This restricted tissue distribution suggests specialized function in pulmonary immunity that warrants further investigation. The a.3 variant lacks the entire EC2 domain, which alters the structural configuration of the receptor and potentially its binding properties . Functionally, these splice variants may exhibit differences in ligand binding affinity, signaling capacity, and cellular distribution. To investigate these variants experimentally, researchers should design PCR primers that span exon junctions to specifically amplify individual splice variants. Western blotting with domain-specific antibodies can confirm protein expression, while functional assays comparing IgA binding capacity, signaling activation, and cellular responses between variants would elucidate their biological significance. Understanding these splice variants is crucial for comprehensive experimental design, as their differential expression might confound results if not properly accounted for.

How does the interaction between FCAR/CD89 and FcR gamma signaling subunit regulate immune responses?

The interaction between FCAR/CD89 and the FcR gamma signaling subunit represents a critical regulatory mechanism that determines the functional outcomes of IgA binding. This interaction is mediated through an electrostatic association between Arg230 in the transmembrane domain of FCAR and a complementary aspartic acid residue in the FcR gamma chain . Upon ligand binding, this association facilitates signal transduction through the immunoreceptor tyrosine-based activation motif (ITAM) present in the FcR gamma chain, leading to downstream activation of Src and Syk family kinases. Interestingly, neutrophils express a significant proportion of FCAR that lacks association with FcR gamma, yet these receptors remain capable of undergoing endocytosis and recycling between early endosomes and the cell surface . To investigate this interaction experimentally, researchers can employ site-directed mutagenesis of the critical Arg230 residue to disrupt FCAR-FcR gamma association and assess the impact on signaling. Co-immunoprecipitation assays can quantify the stoichiometry of FCAR-FcR gamma complexes under different stimulation conditions. Functional assays measuring calcium flux, cytokine production, phagocytosis, and antibody-dependent cell-mediated cytotoxicity (ADCC) should be performed in parallel to correlate receptor-adaptor association with biological outcomes. Advanced imaging techniques such as FRET or proximity ligation assays can provide spatial and temporal information about this interaction in living cells.

What methodological approaches can resolve contradictory data on FCAR/CD89 shedding and circulating immune complexes?

The literature reports conflicting findings regarding FCAR/CD89 shedding and the presence of circulating polymeric IgA/FCAR immune complexes . To resolve these contradictions, researchers should implement a multi-faceted methodological approach. First, standardized sample collection protocols must be established, as proteolytic shedding can occur ex vivo during processing, potentially creating artifacts. Immediate addition of protease inhibitors and careful temperature control during sample handling are essential. Second, a combination of detection methods should be employed, including sensitive ELISAs with antibodies targeting different FCAR epitopes, size-exclusion chromatography followed by Western blotting, and mass spectrometry to definitively identify FCAR fragments. Third, researchers should distinguish between active receptor shedding and the release of membrane vesicles (e.g., microvesicles or exosomes) containing intact FCAR by performing differential ultracentrifugation and vesicle characterization. Fourth, longitudinal studies in well-defined patient cohorts could help determine whether FCAR shedding correlates with disease activity or specific immunological states. Finally, mechanistic studies using inhibitors of different proteases (e.g., matrix metalloproteinases, ADAMs) can identify the enzymes responsible for FCAR shedding. This comprehensive approach would help reconcile contradictory findings and establish whether FCAR shedding represents a physiological regulatory mechanism or a pathological consequence of inflammation.

How can researchers effectively analyze FCAR/CD89 binding kinetics to different IgA forms?

The analysis of FCAR/CD89 binding kinetics to different forms of IgA requires sophisticated biophysical methods that provide quantitative measurements of affinity and binding dynamics. Surface plasmon resonance (SPR) represents the gold standard for such analyses, allowing real-time monitoring of association and dissociation rates between purified FCAR ectodomains and monomeric, dimeric, or secretory IgA . For experimental setup, researchers should generate recombinant FCAR extracellular domains (Gln22-Lys287) with a C-terminal affinity tag for oriented immobilization on sensor chips. Alternatively, IgA forms can be immobilized while FCAR is used as the analyte. Bio-layer interferometry (BLI) provides a complementary approach that requires less sample and is less sensitive to buffer effects. For cell-based binding assays, flow cytometry with fluorescently labeled IgA allows measurement of binding to membrane-expressed FCAR, which maintains its native conformation and associated molecules. Isothermal titration calorimetry (ITC) can provide additional thermodynamic parameters (ΔH, ΔS) to characterize the binding interaction. To ensure accuracy, researchers must account for FCAR glycosylation, which varies depending on the expression system used, as glycans can impact binding affinity. Analysis of binding data should employ appropriate mathematical models that account for the bivalent nature of IgA and the potential for two FCAR molecules to bind a single IgA molecule .

What are the optimal conditions for detecting FCAR/CD89 in flow cytometry experiments?

Optimizing flow cytometry conditions for FCAR/CD89 detection requires careful consideration of several technical parameters. First, antibody selection is critical—validated antibodies like sheep anti-human FCAR/CD89 antigen affinity-purified polyclonal antibody or specific monoclonal antibodies such as clone 488032 conjugated to fluorophores like Alexa Fluor 405 have demonstrated reliability in detecting FCAR . Second, optimal antibody concentration must be determined through titration experiments to identify the concentration that provides maximum specific signal with minimal background. Third, proper sample preparation is essential—use of whole blood rather than isolated cells may better preserve receptor expression, particularly for neutrophils which can undergo rapid activation. Fourth, blocking with appropriate sera (e.g., 10% normal goat serum) prior to antibody staining minimizes non-specific binding. Fifth, inclusion of viability dyes is crucial to exclude dead cells, which can bind antibodies non-specifically. For multicolor panels, careful fluorochrome selection is necessary to minimize spectral overlap with other markers. As shown in validated protocols, staining of human whole blood monocytes and granulocytes with anti-FCAR antibodies followed by appropriate secondary antibodies (if using unconjugated primary) yields distinct positive populations when compared to isotype controls . For quantitative analysis, calibration beads can be used to convert fluorescence intensity to absolute receptor numbers per cell, enabling precise comparison across different experimental conditions.

What strategies can overcome challenges in FCAR/CD89 gene expression analysis?

Gene expression analysis of FCAR presents several challenges that can be addressed through strategic methodological approaches. First, due to the presence of multiple splice variants, primer and probe design is critical—they should either target conserved regions to measure total FCAR expression or span specific exon junctions to distinguish between variants . Second, selection of appropriate reference genes is essential, as traditional housekeeping genes may vary across different myeloid cell populations or activation states. Third, RNA quality assessment is paramount, as neutrophils contain high levels of RNases that can rapidly degrade RNA during isolation. Fourth, normalization to cell-type-specific markers may be necessary when analyzing mixed cell populations. The PrimePCR Probe Assay for FCAR utilizes dual-labeled fluorescent probes that increase specificity and sensitivity compared to SYBR Green-based methods . When using this assay, researchers should follow MIQE guidelines for qPCR experiments, including appropriate controls, standard curves, and melt curve analysis . The assay's amplification plot and standard curve data demonstrate reliable detection across a wide dynamic range (20 to 20 million copies), making it suitable for quantifying FCAR expression in various experimental conditions . For more complex analyses, such as single-cell RNA sequencing, computational approaches must account for the high sequence similarity between FCAR and other Fc receptor family members to ensure accurate read mapping and expression quantification.

How can FCAR/CD89 research findings be integrated into CAR T cell therapy development?

The integration of FCAR/CD89 research findings into CAR T cell therapy development represents an innovative frontier that combines fundamental immunology with translational applications. FCAR's natural ability to recognize IgA-opsonized pathogens and trigger effector functions provides a blueprint for engineering novel chimeric antigen receptors with unique properties . Researchers developing FCAR-based CAR constructs should consider several methodological approaches. First, structure-function studies of FCAR domains can inform the rational design of CAR extracellular regions, potentially incorporating FCAR's EC1 domain for specific recognition properties . Second, the PASCAR (protein abundance structured population dynamic model for CAR T cells) framework can be utilized to predict how different FCAR-derived CAR designs might perform against targets with varying antigen densities . This computational approach allows systematic exploration of design parameters before committing to costly experimental validation. Third, the signaling dynamics of FCAR/FcR gamma interactions can inspire novel intracellular signaling domains for CARs that might provide more physiological activation profiles . Experimental validation should include in vitro cytotoxicity assays against target cells expressing different levels of antigens to assess both efficacy and potential for on-target/off-tumor toxicity . The PASCAR framework has demonstrated the ability to quantitatively describe both in vitro and in vivo results for various CAR designs and can successfully predict experiments outside the training data, making it a valuable tool for FCAR-CAR development .

What approaches should be used to analyze FCAR/CD89 involvement in disease pathogenesis?

Analyzing FCAR/CD89 involvement in disease pathogenesis requires a systematic multi-level approach that integrates genetic, molecular, cellular, and clinical investigations. At the genetic level, researchers should screen for polymorphisms or mutations in the FCAR gene that may correlate with disease susceptibility or progression, using targeted sequencing or genome-wide association studies. At the expression level, quantitative analysis of FCAR transcript and protein levels in patient samples compared to healthy controls can reveal dysregulation patterns . Flow cytometric analysis of receptor density on different myeloid cell populations provides insight into cell-specific alterations . Functional assessments are crucial and should include measuring IgA binding capacity, signaling activation, and effector functions like phagocytosis, oxidative burst, and cytokine production in response to IgA-opsonized targets. Analysis of soluble FCAR and FCAR-IgA immune complexes in patient sera or other biological fluids may reveal disease-specific patterns . For mechanistic studies, animal models expressing human FCAR (as mice lack a direct homolog) can be valuable, though limitations in recapitulating human myeloid biology must be acknowledged. In disease contexts where IgA autoantibodies are present, such as IgA nephropathy or certain autoimmune conditions, investigating how these autoantibodies engage FCAR on myeloid cells could provide critical insights into disease mechanisms. Integration of these datasets using systems biology approaches can ultimately reveal how FCAR contributes to pathogenesis and identify potential points for therapeutic intervention.

Product Science Overview

Introduction

The Fc fragment of IgA receptor, also known as FcαRI or CD89, is a type I transmembrane receptor that plays a crucial role in the immune system. It is primarily expressed on myeloid cells such as neutrophils, eosinophils, monocytes, subsets of dendritic cells, and macrophages . The receptor is responsible for binding the Fc region of Immunoglobulin A (IgA), which is the most abundantly produced antibody isotype in humans .

Structure and Function

The FcαRI receptor is composed of two extracellular immunoglobulin-like domains, a transmembrane domain, and a cytoplasmic tail . The Fc region of IgA, which binds to FcαRI, is derived from the second and third constant domains of the antibody’s heavy chains . This interaction is essential for the receptor-mediated activity of IgA, including its role in immune responses and inflammation .

Role in Immune Response

FcαRI plays a dual role in the immune system. On one hand, it can mediate inhibitory signals that suppress immune responses, thereby preventing the development of autoimmunity and inflammation . On the other hand, when IgA is aggregated, it can induce sustained activation through FcαRI, leading to inflammatory diseases . This Janus-like nature of FcαRI is due to the heterogeneity in molecular forms of IgA and their interaction with the receptor .

Therapeutic Potential

The Fc fragment of IgA receptor has significant therapeutic potential. FcαRI-mediated inhibitory functions have been shown to suppress several inflammatory diseases in animal models, including asthma and glomerulonephritis . Intravenous monomeric IgA (mIgAIV) and anti-FcαR monovalent antibodies are being explored as promising tools for immunotherapy .

Quick Inquiry

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