FCGR3A Human, HEK

CD16a Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Introduction to FCGR3A Human, HEK

FCGR3A (Fc gamma receptor IIIa), also known as CD16a, is a low-affinity receptor for the Fc region of IgG antibodies. The recombinant form "FCGR3A Human, HEK" refers to the human variant of this protein produced in HEK293 (human embryonic kidney) cells . This expression system ensures proper post-translational modifications, critical for functional studies in immunology and therapeutic antibody development.

Functional Roles in Immunity

FCGR3A mediates immune responses via interactions with IgG complexes:

  1. Antibody-Dependent Cellular Cytotoxicity (ADCC):

    • Triggers NK cell activation, leading to lysis of antibody-coated target cells .

    • Polymorphisms (e.g., V158F) influence IgG1/IgG3 binding affinity and ADCC efficacy .

  2. Immune Complex Clearance: Facilitates phagocytosis by macrophages and neutrophils .

  3. Inflammatory Signaling: Activates ITAM-dependent pathways (Syk, PI3K/Akt, NF-κB) upon IgG binding .

Genetic Polymorphisms and Disease Associations

PolymorphismFunctional ImpactClinical Correlation
V158F (rs396991)Higher affinity for IgG1/IgG3 in V158 variant - Improved response to anti-CD20 (rituximab) in lymphoma .
- Increased SLE susceptibility in F158 carriers .
G559TAltered glycosylation and FcγRIIIa stability Linked to recurrent viral infections .

Therapeutic Antibody Interactions

  • Anti-CD20 (Rituximab): FCGR3A-V158 enhances NK cell-mediated ADCC, correlating with prolonged survival in non-Hodgkin’s lymphoma .

  • Anti-EGFR (Cetuximab): FCGR3A genotype affects clinical outcomes in colorectal cancer (HR = 1.59 for V/V vs. F/F) .

Recombinant Production in HEK293 Cells

  • Expression System: HEK293 cells ensure mammalian glycosylation patterns .

  • Purification: Proprietary chromatography (e.g., Ni-NTA for His-tag) .

  • Purity: >95% by SDS-PAGE and HPLC .

Key Applications in Research

  1. ADCC Assays: Quantify NK cell cytotoxicity using anti-CD16 antibodies .

  2. Immune Checkpoint Studies: Investigate FcγRIIIa’s role in tumor microenvironment modulation .

  3. Therapeutic Antibody Development: Optimize IgG Fc engineering for enhanced FcγRIIIa binding .

Challenges and Future Directions

  • Glycosylation Variability: Impacts FcγRIIIa-IgG affinity; requires stringent batch-to-batch monitoring .

  • Allele-Specific Therapies: Targeting FCGR3A polymorphisms (e.g., V158F) could personalize monoclonal antibody treatments .

Product Specs

Introduction
Low affinity immunoglobulin gamma Fc region receptor III-A (FCGR3A) is a receptor responsible for binding to the Fc portion of immunoglobulin G. This interaction plays a crucial role in eliminating antigen-antibody complexes from the circulatory system and participates in other antibody-dependent immune responses. FCGR3A requires association with the gamma subunit of Fc epsilon for its function. Primarily found on natural killer (NK) cells, FCGR3A exists as an integral membrane glycoprotein anchored via a transmembrane peptide. In contrast, FCGR3B is expressed on polymorphonuclear neutrophils (PMN) and anchored through a phosphatidylinositol (PI) linkage. FCGR3A is also found on macrophages, certain T-cell subsets, immature thymocytes, and placental trophoblasts. Its functions include mediating antibody-dependent cellular cytotoxicity (ADCC) and other antibody-dependent processes like phagocytosis. Mutations in the FCGR3A gene have been associated with increased susceptibility to recurrent viral infections, systemic lupus erythematosus, and alloimmune neonatal neutropenia.
Description
Recombinant human FCGR3A, produced in HEK293 cells, is a single polypeptide chain consisting of 197 amino acids (residues 18-208). With a molecular weight of 22.6 kDa, the protein contains a 6-amino acid His-tag fused at the C-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The FCGR3A protein solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of phosphate-buffered saline (pH 7.4) and 10% glycerol.
Stability
For optimal storage, keep the vial at 4°C if it will be used within 2-4 weeks. For longer-term storage, freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid subjecting the solution to multiple freeze-thaw cycles.
Purity
The purity of the FCGR3A protein is greater than 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The biological activity of FCGR3A is assessed by its binding affinity to Human IgG1 Fc in a functional ELISA (using catalog number pro-2763).
Synonyms
Low affinity immunoglobulin gamma Fc region receptor III-A, CD16a antigen, Fc-gamma, RIII-alpha, Fc-gamma RIII, Fc-gamma RIIIa, FcRIII, FcRIIIa, FcR-10, IgG Fc receptor III-2, CD16a, FCGR3A, FCG3, FCGR3, IGFR3, CD16, FCGRIII.
Source

HEK293 Cells

Amino Acid Sequence

MRTEDLPKAV VFLEPQWYRV LEKDSVTLKC QGAYSPEDNS TQWFHNESLI SSQASSYFID AATVDDSGEY RCQTNLSTLS DPVQLEVHIG WLLLQAPRWV FKEEDPIHLR CHSWKNTALH KVTYLQNGKG RKYFHHNSDF YIPKATLKDS GSYFCRGLFG SKNVSSETVN ITITQGLAVS TISSFFPPGY Q HHHHHH.

Q&A

What is FCGR3A and what are its key molecular features?

FCGR3A (Low affinity immunoglobulin gamma Fc region receptor III-A) is a receptor for the Fc portion of immunoglobulin G that plays a crucial role in immune responses. The human recombinant FCGR3A produced in HEK293 cells is a single polypeptide chain containing 197 amino acids (positions 18-208) with a molecular mass of 22.6 kDa, typically fused to a 6-amino acid His-tag at the C-terminus for purification purposes . This receptor is involved in the elimination of antigen-antibody complexes from circulation and mediates various antibody-dependent responses, including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis . FCGR3A requires association with the gamma subunit of Fc epsilon receptor for optimal function .

How does FCGR3A differ from other Fc gamma receptors, particularly FCGR3B?

While FCGR3A and FCGR3B share significant sequence homology, they differ in their cellular expression patterns and membrane anchoring mechanisms. FCGR3A is expressed on natural killer (NK) cells as an integral membrane glycoprotein anchored through a transmembrane peptide, whereas FCGR3B is expressed on polymorphonuclear neutrophils (PMN) and is anchored through a phosphatidylinositol (PI) linkage . Additionally, FCGR3A has broader expression than FCGR3B, being found on macrophages, subpopulations of T-cells, immature thymocytes, and placental trophoblasts . Functionally, FCGR3A mediates antibody-dependent cellular cytotoxicity and triggers lysis of target cells independently of IgG binding in some contexts .

What is the cellular distribution pattern of FCGR3A?

FCGR3A exhibits a distinct cellular distribution pattern critical to its immune functions. It is predominantly expressed on natural killer (NK) cells as an integral membrane glycoprotein anchored through a transmembrane peptide . Additionally, FCGR3A expression has been documented on macrophages, subpopulations of T-cells, immature thymocytes, and placental trophoblasts . This distribution pattern differs from FCGR3B, which is primarily expressed on polymorphonuclear neutrophils . Understanding this distribution pattern is essential for designing experiments that accurately reflect physiological conditions in which FCGR3A functions.

Why are HEK293 cells preferred for FCGR3A recombinant protein production?

HEK293 cells represent a preferred expression system for FCGR3A recombinant protein production due to their mammalian origin, which ensures proper post-translational modifications crucial for FCGR3A functionality. As a human-derived cell line, HEK293 cells provide appropriate glycosylation patterns that can significantly impact the receptor's binding affinity and biological activity . The human cellular machinery in HEK293 cells facilitates proper folding and processing of complex proteins like FCGR3A, which contains multiple disulfide bonds and glycosylation sites. This expression system yields recombinant FCGR3A that more closely resembles the native protein structure compared to bacterial or insect cell expression systems, making it particularly valuable for functional studies and therapeutic applications requiring high biological fidelity.

What are the optimal purification strategies for FCGR3A expressed in HEK293 cells?

Purification of FCGR3A from HEK293 cells typically employs a multi-step chromatographic approach. The recombinant protein is commonly designed with a C-terminal 6-amino acid His-tag to facilitate purification using immobilized metal affinity chromatography (IMAC) . After initial capture using IMAC, additional purification steps may include ion exchange chromatography to separate charged variants and size exclusion chromatography to ensure monomeric purity and remove aggregates. These proprietary chromatographic techniques yield high-purity FCGR3A suitable for functional and structural studies . For optimal results, purification should be performed rapidly at controlled temperatures (typically 4°C) to minimize protein degradation, and protease inhibitors should be included in early purification stages to prevent proteolytic cleavage.

What formulation conditions maintain optimal stability of recombinant FCGR3A?

The stability of recombinant FCGR3A depends critically on appropriate formulation conditions. The protein solution (typically at 0.5mg/ml) is optimally maintained in a formulation containing phosphate-buffered saline (pH 7.4) with 10% glycerol . This formulation provides an isotonic environment at physiological pH while the glycerol component acts as a cryoprotectant and stabilizing agent. For short-term storage (2-4 weeks), the protein can be stored at 4°C, while longer-term storage requires freezing at -20°C . To prevent freeze-thaw degradation, it is advisable to aliquot the protein before freezing. For extended storage periods, the addition of a carrier protein (such as bovine serum albumin at 0.1-1%) is recommended to prevent surface adsorption and maintain activity . These conditions collectively minimize protein denaturation, aggregation, and loss of functional activity.

What are the established assays for evaluating FCGR3A binding affinity and functional activity?

Several validated assays exist for characterizing FCGR3A binding and functional properties. Surface plasmon resonance (SPR) represents a gold-standard approach for quantifying binding affinities between FCGR3A and various IgG subtypes or Fc-engineered antibodies . Reporter cell assays utilizing BW5147 cells expressing human FcγRIIIA as a chimeric molecule with mouse CD3ξ have been developed to measure receptor activation, where activation leads to mIL-2 secretion quantifiable by ELISA . For evaluating antibody-dependent cellular cytotoxicity (ADCC), NK cell degranulation assays measuring CD107a positivity provide a physiologically relevant readout . Additionally, flow cytometry-based approaches can assess competitive binding of FCGR3A to immune complexes or opsonized targets . When evaluating polymorphic variants, high-throughput FCGR genotyping using the Illumina MiSeq platform enables precise allelic discrimination . These methodologies collectively provide comprehensive characterization of FCGR3A's binding properties and functional activities in various experimental contexts.

How can researchers investigate FCGR3A-mediated antibody-dependent cellular cytotoxicity (ADCC)?

Investigation of FCGR3A-mediated ADCC requires carefully designed experimental approaches that recapitulate physiological conditions. Primary NK cell degranulation assays represent a direct method where peripheral blood mononuclear cells (PBMCs) are co-cultured with antibody-opsonized target cells, and CD107a surface expression is measured as a marker of degranulation . For higher throughput analysis, reporter cell systems using FcγRIIIA-expressing BW5147 cells that produce IL-2 upon receptor activation can be employed . The specific antibody concentration is critical, as optimal ADCC typically occurs at antibody concentrations reflecting the plateau of the dose-response curve. When evaluating therapeutic antibodies, comparison between wild-type and Fc-engineered variants can reveal enhancements in ADCC potential, with afucosylated IgGs demonstrating enhanced ADCC activity through FCGR3A . Researchers should consider the ratio of effector to target cells, antibody concentration, and incubation time as key variables that influence assay sensitivity and reproducibility.

What are the implications of FCGR3A polymorphisms for research design and data interpretation?

FCGR3A exhibits significant genetic polymorphism with functional consequences that must be considered in experimental design. The FCGR3A F158V polymorphism has been associated with differential clinical outcomes, including decreased risk for bloodstream infections following liver transplantation . This polymorphism affects the receptor's binding affinity for IgG, with the V158 variant demonstrating higher affinity. When designing studies, researchers should genotype FCGR3A in primary cells or select cell lines with known polymorphic status to ensure reproducibility . High-throughput FCGR genotyping methods using the Illumina MiSeq platform can efficiently characterize experimental samples . Understanding FCGR3A haplotypes is also critical, as they can influence receptor expression levels and function. Data interpretation should account for these polymorphic differences, particularly when comparing results across different donor samples or cell lines. Stratification of experimental data by FCGR3A genotype may reveal important associations that would otherwise be masked in pooled analyses.

How do FCGR3A gene mutations contribute to immune-related disorders?

FCGR3A gene mutations have significant implications for immune system function and disease susceptibility. These mutations are linked to increased susceptibility to recurrent viral infections, systemic lupus erythematosus, and alloimmune neonatal neutropenia . The mechanisms underlying these associations involve altered binding affinity for IgG, modified signal transduction, or disrupted expression patterns. In systemic lupus erythematosus, FCGR3A mutations may impair clearance of immune complexes, contributing to tissue deposition and inflammation. For viral infections, compromised FCGR3A function potentially reduces antibody-dependent cellular cytotoxicity (ADCC) against virus-infected cells. In alloimmune neonatal neutropenia, maternal antibodies against paternal FCGR3A variants can lead to destruction of fetal neutrophils. Comprehensive genetic analysis using next-generation sequencing approaches can identify specific mutations in research or clinical contexts, providing insights into pathogenic mechanisms and potential therapeutic targets for these immune-related disorders.

What is the role of FCGR3A in viral defense mechanisms, particularly against human cytomegalovirus?

FCGR3A plays a critical role in antiviral immunity, particularly evident in the complex interactions with human cytomegalovirus (HCMV). This receptor mediates antibody-dependent cellular cytotoxicity (ADCC) against virus-infected cells when they are opsonized with virus-specific antibodies . HCMV has evolved sophisticated countermeasures to evade this immune surveillance, expressing viral Fcγ receptor homologs (vFcγRs) such as gp34 and gp68 that antagonize FCGR3A activation . These viral proteins work synergistically to disrupt FcγR binding to antibody-opsonized infected cells, with gp68 directly interfering with FCGR3A binding to immune complexes . Experimental systems utilizing reporter cells expressing human FcγRIIIA have demonstrated approximately 60% reduction in FcγRIII binding in the presence of gp68 . This viral evasion strategy highlights the evolutionary importance of FCGR3A in controlling HCMV infection and suggests potential therapeutic approaches targeting these viral antagonists to enhance antiviral immunity.

How does FCGR3A contribute to the efficacy of therapeutic monoclonal antibodies?

FCGR3A significantly influences the clinical efficacy of therapeutic monoclonal antibodies through its role in mediating antibody-dependent cellular cytotoxicity (ADCC). Upon binding to antibody-coated target cells, FCGR3A triggers TNFA-dependent ADCC, particularly important for the antitumor activities of therapeutic antibodies . The receptor's polymorphic variants affect this process, with the V158 variant demonstrating higher binding affinity to IgG and potentially enhanced ADCC compared to the F158 variant. This genetic variation partly explains differential patient responses to monoclonal antibody therapies . Engineering antibodies with modified Fc regions that enhance FCGR3A binding can improve therapeutic efficacy, as demonstrated by afucosylated IgGs that promote enhanced ADCC through this receptor . When designing or evaluating therapeutic antibodies, researchers should consider FCGR3A polymorphism status in preclinical models and clinical studies to accurately predict efficacy across genetically diverse populations. This mechanistic understanding has driven the development of next-generation therapeutic antibodies with optimized Fc domains for maximal FCGR3A engagement.

What are the emerging technologies for studying FCGR3A interactions with engineered antibodies?

Advanced technologies are revolutionizing the study of FCGR3A interactions with engineered antibodies. High-throughput surface plasmon resonance (SPR) platforms enable systematic evaluation of binding affinities between FCGR3A variants and diverse antibody Fc modifications . Single-molecule imaging techniques can visualize receptor clustering and signaling dynamics in real-time following antibody engagement. CRISPR-Cas9 gene editing allows precise modification of FCGR3A in cell lines to study structure-function relationships. For more complex analyses, integrated approaches combining crystallography or cryo-electron microscopy with molecular dynamics simulations provide atomic-level insights into binding interfaces and conformational changes. Biocomputational methods leveraging machine learning can predict optimal antibody Fc modifications for enhanced FCGR3A engagement. These methodological advances collectively enable more sophisticated understanding of FCGR3A-antibody interactions, facilitating rational design of next-generation therapeutic antibodies with precisely tailored effector functions and improved clinical outcomes for patients with diverse FCGR3A genotypes.

What are the key considerations when designing experiments to study viral evasion of FCGR3A-mediated immunity?

Designing robust experiments to investigate viral evasion of FCGR3A-mediated immunity requires careful consideration of multiple variables. Researchers must select appropriate viral strains, as genetic differences between laboratory-adapted and clinical isolates may affect immune evasion mechanisms . When studying human cytomegalovirus (HCMV), the expression of viral Fcγ receptor homologs (vFcγRs) like gp34 and gp68 should be verified, as these proteins synergistically antagonize FCGR3A activation . The antibody source is critical; using hyperimmunoglobulin preparations with abundant non-immune IgG (like Cytotect) more accurately reflects physiological conditions than monoclonal antibodies alone . Reporter cell systems expressing chimeric FcγRIIIA provide quantifiable readouts through IL-2 secretion, while primary NK cell degranulation assays (measuring CD107a positivity) offer physiologically relevant endpoints . Importantly, the ratio of non-immune to immune IgG significantly influences vFcγR antagonism, with optimal synergism between gp34 and gp68 observed at a 100:1 ratio . Control experiments should include viruses lacking specific immune evasion genes and blockade of FCGR3A to confirm specificity. Time-course analyses are essential, as viral evasion mechanisms may show temporal regulation during infection.

Product Science Overview

Expression and Structure

CD16a is encoded by the FCGR3A gene and is expressed on the surface of several immune cells, including natural killer (NK) cells, macrophages, a subset of T-cells, immature thymocytes, and placental trophoblasts . It is a transmembrane glycoprotein that is anchored to the cell membrane via a polypeptide chain. The receptor has an intermediate affinity for IgG antibodies, which allows it to bind to the Fc region of these antibodies and mediate immune responses .

The recombinant form of CD16a is often expressed in HEK 293 cells (Human Embryonic Kidney 293 cells), which are widely used in biological research due to their high transfection efficiency and reliable protein expression . The recombinant protein is typically tagged with a 6-His tag at the C-terminus to facilitate purification and detection .

Functional Significance

CD16a is involved in several critical immune functions:

  1. Phagocytosis: CD16a mediates the engulfment and digestion of pathogens and cellular debris by immune cells.
  2. Secretion of Enzymes and Inflammatory Mediators: Upon activation, CD16a triggers the release of enzymes and inflammatory molecules that help in the immune response.
  3. Antibody-Dependent Cellular Cytotoxicity (ADCC): CD16a plays a key role in ADCC, where NK cells recognize and kill antibody-coated target cells.
  4. Clearance of Immune Complexes: CD16a helps in the removal of immune complexes from the circulation, preventing their deposition in tissues and subsequent inflammation .
Clinical Relevance

Aberrant expression or mutations in CD16a have been implicated in various diseases and conditions, including:

  • Recurrent Viral Infections: Defective CD16a function can lead to increased susceptibility to viral infections.
  • Systemic Lupus Erythematosus (SLE): Altered CD16a expression is associated with the autoimmune disease SLE.
  • Alloimmune Neonatal Neutropenia: Mutations in CD16a can cause this condition, characterized by low levels of neutrophils in newborns .
Recombinant CD16a in Research

Recombinant CD16a expressed in HEK 293 cells is widely used in research to study its structure, function, and role in immune responses. The recombinant protein is typically lyophilized from a filtered solution in phosphate-buffered saline (PBS) with added protectants like mannitol or trehalose . It can be reconstituted in sterile PBS for experimental use and stored at -20°C for extended periods .

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.