FCGR2A Human, HEK refers to recombinant human Fc gamma receptor IIa (CD32a) produced in HEK-293 cell lines. These cells are engineered to express the extracellular domain of FCGR2A (typically residues 36–218), often fused with tags like His-Avi for purification and detection . Key features include:
Purity: >95% monomer purity verified by SDS-PAGE and SEC-HPLC .
Allotypes: Two polymorphic variants exist at position 167:
IgG Binding Specificity:
Regulatory Elements: CRISPRi screens identified six genomic regions regulating FCGR2A expression, including a proximal enhancer in intron 3 and CTCF-bound distal enhancers .
Autoimmunity: The H167 variant correlates with SLE susceptibility due to enhanced IgG2 immune complex clearance .
Infectious Diseases: H167 improves phagocytosis of IgG2-opsonized pathogens (e.g., Streptococcus pneumoniae) .
Therapeutic Response:
HEK-derived FCGR2A proteins are pivotal for:
Binding Assays: Quantifying antibody-Fc interactions via SPR (e.g., trastuzumab KD = 1.7 μM ; rituximab KD = 1.23 μM ).
Structural Studies: Mapping epitopes for therapeutic antibody optimization .
Drug Screening: Evaluating Fc-engineered biologics targeting autoimmune or oncologic pathways .
Recombinant human FCGR2A is a glycosylated polypeptide chain encompassing amino acids 30 to 218, resulting in a total of 195 amino acids. Its molecular mass is 21.6 kDa. For purification purposes, a 6-amino acid His-tag is fused to the C-terminus, and proprietary chromatographic techniques are employed.
The FCGR2A protein is provided at a concentration of 1 mg/ml in a solution containing 10% glycerol and phosphate-buffered saline (pH 7.4).
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended periods, storage at -20°C in a frozen state is recommended. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Multiple freeze-thaw cycles should be avoided.
The purity of the product is greater than 95.0%, as determined by SDS-PAGE analysis.
The ED50 value, determined through a functional ELISA utilizing Human IgG1 Fc (CAT# pro-2763), is less than or equal to 5 ug/ml, indicating its binding capability.
CD32, CD32A, CDw32, FcGR, IGFR2, MGC23887, MGC30032, Low affinity immunoglobulin gamma Fc region receptor II-a, IgG Fc receptor II-a, Fc-gamma-RIIa, FcRII-a, FCG2, FCGR2A1.
HEK293 Cells.
SADSQAAAPP KAVLKLEPPW INVLQEDSVT LTCQGARSPE SDSIQWFHNG NLIPTHTQPS YRFKANNNDS GEYTCQTGQT SLSDPVHLTV LSEWLVLQTP HLEFQEGETI MLRCHSWKDK PLVKVTFFQN GKSQKFSHLD PTFSIPQANH SHSGDYHCTG NIGYTLFSSK PVTITVQVPS MGSSSPMGIH HHHHH.
FCGR2A (CD32a) is a low-affinity receptor for the Fc fragment of IgG that binds antibody-antigen complexes to regulate the abundance of circulating and deposited immune complexes. This receptor plays a critical role in modulating downstream immune and autoimmune responses by mediating interactions between immune cells and antibody-coated targets . FCGR2A is widely expressed on hematopoietic cells and serves as an important link between humoral and cellular immunity, participating in both innate and adaptive immune responses to IgG immune complexes .
Recombinant FCGR2A proteins expressed in HEK-293 cells typically include the extracellular domain (from approximately Ala36-Ile218) of the receptor. These proteins can be engineered with various purification or detection tags, including His tags, Fc tags, or Avi tags for biotinylation . The recombinant proteins can achieve high purity (>95% as determined by methods like SDS-PAGE and HPLC) with minimal endotoxin contamination (<0.1EU/μg) . When properly folded, these proteins maintain their biological activity and antibody-binding capabilities, making them valuable tools for studying receptor-ligand interactions in controlled experimental settings.
The FCGR2A gene has a common non-synonymous single nucleotide polymorphism (SNP) that results in either histidine (H) or arginine (R) at position 131, corresponding to position 167 in some numbering systems. The H131 variant (CD32a H) binds human IgG1 immune complexes more avidly than the R131 variant (CD32a R) . This differential binding affinity has important biological consequences, as the H131 variant is strongly associated with several autoimmune diseases including rheumatoid arthritis and inflammatory bowel disease . Recent research has shown that the H131 variant more readily forms a ternary complex with the neonatal Fc receptor (FcRn) under acidic conditions, which enhances immune responses to IgG immune complexes .
CRISPR-based technologies offer powerful approaches for investigating FCGR2A expression regulation. Two complementary methods have proven particularly useful: CRISPR interference (CRISPRi) and standard CRISPR/Cas9 cutting. CRISPRi employs an enzymatically inactive Cas9 (dCas9) fused to the Krüppel associated box (KRAB) domain to induce histone deacetylation and tri-methylation at histone 3 leucine 9 (H3K9me3), effectively silencing regulatory regions . This approach can identify enhancers and promoters affecting FCGR2A expression across large genomic regions. For higher resolution mapping, standard CRISPR/Cas9 cutting with guide RNAs tiled across potential regulatory elements can precisely define functional regulatory subregions . Together, these techniques have identified specific regulatory elements both upstream of the transcription start site and within intron 3 of FCGR2A that significantly influence its expression levels.
When designing experiments to assess FCGR2A binding affinity to various ligands, several key parameters must be considered. Functional ELISA assays have been used successfully to measure binding interactions, with immobilized Human FCGR2A/CD32a(R167) at concentrations of approximately 1 μg/mL (100 μL/well) . Under these conditions, binding interactions with specific antibodies can be detected within a linear range of approximately 0.977-175.35 ng/mL . For studying interactions with immune complexes, it's important to note that pH can significantly affect binding properties, particularly when investigating interactions with other Fc receptors like FcRn. Acidic conditions have been shown to facilitate formation of ternary complexes between FCGR2A, IgG, and FcRn . Flow cytometry-based assays also provide valuable tools for quantifying receptor surface expression and binding in both cell lines and primary human cells.
Topologically associated domains (TADs) provide crucial information for comprehensively studying FCGR2A regulatory elements. Hi-C data from relevant cell lines like K562 (erythroleukemia) and THP-1 (acute monocytic leukemia) have revealed that FCGR2A resides within a defined TAD of approximately 400 kb (chr1:161,400,000-161,800,000) . This genomic architecture helps explain how distal regulatory elements may influence FCGR2A expression despite being located far from the gene itself. When designing sgRNA libraries for regulatory element screening, it's advisable to cover the entire TAD containing FCGR2A and potentially adjacent TADs to capture all relevant regulatory interactions. Recent studies have successfully employed this approach by designing lentiviral libraries of over 22,000 sgRNAs targeting all regions of open chromatin within defined TADs surrounding FCGR2A . This comprehensive approach enables systematic identification of both proximal and distal regulatory elements controlling FCGR2A expression.
Successful expression of FCGR2A variants in HEK-293 cells requires careful optimization of several parameters. The expression construct design is crucial - typically incorporating the extracellular domain of FCGR2A (approximately Ala36-Ile218) with appropriate fusion tags for purification and detection . For investigating variant-specific effects, precise introduction of the desired polymorphism (such as His167Arg) is essential. Transfection methods must be optimized for HEK-293 cells, with transient transfection systems often yielding good results for research-scale production . Post-translational modifications, particularly glycosylation patterns, can significantly affect receptor function, so culture conditions should be standardized to ensure consistent glycoform distributions. Serum-free media formulations are generally preferred to avoid contamination with bovine immunoglobulins that might interfere with receptor function assessments. Expression levels should be monitored by methods such as flow cytometry or Western blotting to confirm successful production before proceeding to purification steps.
Multiple purification strategies can be employed to obtain high-quality FCGR2A protein preparations, with the optimal approach depending on the specific fusion tags incorporated into the expression construct. For His-tagged constructs, immobilized metal affinity chromatography (IMAC) using Ni-NTA or similar resins provides efficient initial capture . For Fc-tagged constructs, protein A or protein G affinity chromatography offers high selectivity. When higher purity is required, a multi-step purification process is recommended, typically combining initial affinity purification with subsequent size exclusion chromatography (SEC) to remove aggregates and achieve monodisperse preparations. Quality control analyses should include SDS-PAGE (>95% purity), SEC-HPLC (monodispersity assessment), endotoxin testing (<0.1EU/μg), and functional validation through binding assays . Sterile filtration (0.22 μm) is essential for preparing samples for cell-based assays. For biotinylated constructs, especially those with Avi tags, enzymatic biotinylation either in vivo during expression or in vitro post-purification must be carefully controlled and quantified to ensure consistent functional properties .
Verifying the functional activity of purified FCGR2A proteins is essential before using them in complex experimental systems. Functional ELISA represents a primary validation method, where immobilized FCGR2A protein (typically at 1 μg/mL) is assessed for its ability to bind specific antibodies or immune complexes across a range of concentrations . Surface plasmon resonance (SPR) provides quantitative binding kinetics and affinity constants, offering deeper insights into receptor-ligand interactions. Flow cytometry-based competition assays, where the purified protein competes with cellular FCGR2A for ligand binding, can confirm native-like binding properties. For dimeric constructs, such as those utilizing an Fc fusion strategy, analytical ultracentrifugation or multi-angle light scattering can verify the correct oligomeric state . When studying variant-specific effects (e.g., His167Arg), comparative binding assays should demonstrate the expected differences in affinity for IgG subtypes. Finally, cell-based functional assays measuring immune complex uptake or cellular activation in the presence of blocking concentrations of purified receptor provide the most physiologically relevant validation of functional integrity.
The FCGR2A H131 variant demonstrates enhanced interaction capabilities with FcRn compared to the R131 variant. Under acidic conditions, the H131 variant more readily forms a ternary complex with FcRn and IgG immune complexes . This interaction appears to be functionally significant, as FcRn serves as a co-receptor for CD32a in mediating responses to IgG immune complexes. In cellular systems, both CD32a variants require FcRn to induce optimal innate and adaptive immune responses to human IgG1 immune complexes, but these responses are significantly augmented in the presence of the H131 variant . These molecular interactions provide a mechanistic explanation for the association between the H131 variant and autoimmune diseases. When designing experiments to investigate these interactions, researchers should carefully control pH conditions, as the CD32a-FcRn interaction is pH-dependent, showing stronger association under acidic conditions that mimic the endosomal environment where these receptors typically function together.
Detecting functional differences between FCGR2A variants requires carefully designed experimental approaches that are sensitive to the specific binding and signaling properties of each variant. Flow cytometry using variant-specific antibodies or genetically defined primary cells provides a direct method for quantifying surface expression levels in different cellular contexts . For binding studies, surface plasmon resonance (SPR) with purified receptors and various IgG subtypes or immune complexes can quantitatively measure affinity differences between variants. Cell-based functional assays measuring phagocytosis, antibody-dependent cellular cytotoxicity (ADCC), or cytokine production in response to immune complexes offer physiologically relevant readouts of variant-specific activity. Genetics-based approaches, including CRISPR-engineered cell lines expressing specific variants, provide clean systems for comparative studies. Additionally, affinity capture mass spectrometry (AC-MS) assays have been developed to specifically distinguish between FcγRIIa allotypes, offering a powerful tool for studying antibody-dependent cellular phagocytosis (ADCP) and other functional endpoints .
Modeling the impact of FCGR2A polymorphisms in disease-relevant systems requires thoughtfully designed experimental platforms that recapitulate key aspects of disease pathophysiology. Patient-derived cells genotyped for FCGR2A variants provide the most directly relevant system, though variability between donors can complicate interpretation. CRISPR-engineered isogenic cell lines expressing different FCGR2A variants offer controlled systems for mechanistic studies, eliminating confounding genetic factors. For autoimmune disease models, co-culture systems incorporating relevant immune cell types (monocytes, neutrophils, B cells) can assess how FCGR2A variants affect cellular cross-talk in the presence of disease-specific immune complexes . Humanized mouse models expressing human FCGR2A variants permit in vivo assessment of variant-specific effects on disease progression, particularly in models of rheumatoid arthritis or other autoimmune conditions associated with the H131 variant . When designing these systems, researchers should consider that optimal immune responses to IgG immune complexes require both FCGR2A and FcRn, with the H131 variant showing enhanced co-receptor function with FcRn . This suggests that therapeutic approaches targeting this interaction, rather than merely reducing autoantibody levels, may offer novel treatment strategies for autoimmune diseases associated with the H131 variant.
Systematically identifying genomic elements regulating FCGR2A expression requires a multi-faceted approach combining various genomic technologies. CRISPR interference (CRISPRi) with dCas9-KRAB provides an efficient screening platform for identifying both proximal and distal regulatory elements . This approach can be implemented by designing comprehensive sgRNA libraries targeting regions of open chromatin within the topologically associated domains (TADs) surrounding FCGR2A. Following CRISPRi screening, high-resolution mapping of identified regulatory regions can be achieved using standard CRISPR/Cas9 cutting with densely tiled sgRNAs . Integration of these functional screens with epigenomic data, including ChIP-seq for histone modifications (H3K4me1, H3K27Ac) and ATAC-seq for chromatin accessibility, helps prioritize biologically relevant regulatory elements . For even deeper characterization, chromosome conformation capture techniques (3C, 4C, Hi-C) can identify long-range interactions between the FCGR2A promoter and distal regulatory elements. This systematic approach has successfully identified key regulatory regions affecting FCGR2A expression, including both activating and inhibitory elements located in the promoter region and within intron 3 .
Identification of transcription factors (TFs) involved in FCGR2A expression regulation can be accomplished through several complementary approaches. ChIP-seq experiments targeting specific TFs can directly identify binding sites within the regulatory regions previously identified through CRISPRi and CRISPR/Cas9 screening . For broader discovery, motif analysis of these regulatory regions can predict potential TF binding sites, which can then be validated through functional assays. The most critical transcription factor binding sites appear to be concentrated in two subregions: one within 250 bp upstream of the transcription start site (TSS) and another in intron 3, particularly within 40 bp of the exon 3 junction . These regions demonstrate high chromatin accessibility in both cell lines and primary monocytes. Interestingly, within intron 3, there appears to be a complex regulatory landscape with some elements enhancing and others repressing FCGR2A expression . When designing experiments to study these transcription factors, researchers should consider that inflammation-associated TFs may be particularly relevant, given FCGR2A's role in immune function and the association of certain variants with autoimmune diseases.
Genetic variants within regulatory regions can significantly impact FCGR2A expression through various mechanisms. Luciferase reporter assays using constructs containing wild-type and variant sequences of identified regulatory elements provide a direct method for quantifying their effect on transcriptional activity . These effects can be validated in genotyped individuals using flow cytometry to measure surface expression levels of FCGR2A protein . Variants may disrupt or create binding sites for specific transcription factors, which can be assessed through electrophoretic mobility shift assays (EMSA) or DNA-protein interaction ELISA. For a mechanistic understanding, CRISPR base editing to introduce specific variants into regulatory regions of otherwise isogenic cells allows precise determination of variant effects. Integration of expression quantitative trait loci (eQTL) data from large cohorts with FCGR2A genotyping can further connect genetic variation to expression differences at the population level. Given the association of FCGR2A variants with autoimmune diseases, studying how regulatory variants interact with coding variants (such as His167Arg) may reveal compound effects that contribute to disease susceptibility or severity through both expression level changes and functional differences in the receptor protein.
FCGR2A expression systems provide valuable tools for optimizing therapeutic antibody development. By expressing both FCGR2A variants (H131 and R131) in controlled cell systems like HEK-293, researchers can systematically evaluate how candidate therapeutic antibodies interact with these receptors . This is particularly important for antibodies targeting diseases where the FCGR2A genotype affects pathology or treatment response. High-throughput screening platforms incorporating these expression systems can identify antibody modifications that enhance or reduce FCGR2A binding, depending on the desired therapeutic mechanism. For diseases associated with the H131 variant, such as rheumatoid arthritis, understanding how therapeutic antibodies interact differently with each variant could enable personalized medicine approaches . Additionally, the discovery that FCGR2A forms functional complexes with FcRn suggests that therapeutic strategies might target this interaction rather than simply focusing on reducing autoantibody levels . Affinity capture mass spectrometry (AC-MS) assays specifically developed for different FCGR2A allotypes provide powerful tools for characterizing antibody-dependent cellular phagocytosis (ADCP) activities of therapeutic antibody candidates .
The FCGR2A H131 (His167) polymorphism has significant implications for predicting autoimmune disease susceptibility. This variant binds human IgG1 immune complexes more avidly than the R131 variant and forms ternary complexes with FcRn under acidic conditions . These molecular interactions translate to enhanced immune and inflammatory responses to IgG immune complexes, potentially explaining the strong association between the H131 variant and several autoimmune diseases, including rheumatoid arthritis and inflammatory bowel disease . Large-scale genetic association studies have consistently identified this polymorphism as a risk factor, though the effect size varies across different populations and disease subtypes. For clinical applications, genotyping patients for FCGR2A variants may help stratify disease risk, particularly in individuals with other genetic or environmental risk factors. Furthermore, understanding the functional consequences of this polymorphism informs the development of targeted therapeutics. Recent research demonstrating that FcRn blockade can decrease inflammation in rheumatoid arthritis models without reducing circulating autoantibody levels suggests novel intervention strategies targeting the FCGR2A-FcRn interaction rather than antibody levels alone .
Integrating FCGR2A expression systems with studies of other immune receptors provides a more comprehensive understanding of immune complex processing and inflammatory responses. Co-expression systems incorporating FCGR2A variants alongside FcRn have revealed crucial functional interactions between these receptors in mediating responses to IgG immune complexes . Similar approaches can be applied to study interactions with other Fc receptors (FcγRI, FcγRIIb, FcγRIII) that may cooperate or compete with FCGR2A in immune complex recognition. For mechanistic studies, CRISPR-engineered cell lines expressing fluorescently tagged versions of multiple receptors enable real-time imaging of receptor clustering, trafficking, and signal integration. Multi-parametric flow cytometry or mass cytometry (CyTOF) with cells expressing defined receptor variants allows quantification of complex signaling networks across diverse immune cell populations. When applying these integrated systems to disease models, researchers should consider that optimal responses to IgG immune complexes require both classical FcγRs and atypical receptors like FcRn, with specific variants (like FCGR2A H131) showing enhanced cooperation . This systems-level approach to receptor interactions may reveal novel therapeutic targets at the interface between different receptor families, potentially offering more selective intervention strategies for autoimmune diseases associated with aberrant immune complex processing.
CD32a is a cell surface receptor predominantly expressed on phagocytic cells such as macrophages and neutrophils . The gene encoding CD32a is part of a family of genes that includes other Fc receptors like FcγRIIb and FcγRIIc. These receptors are encoded by mRNA splice variants of three highly related genes: FCGR2A, FCGR2B, and FCGR2C .
The recombinant form of CD32a, expressed in HEK293 cells (Human Embryonic Kidney 293 cells), is often used in research to study its function and interactions. The recombinant protein typically includes a His-tag for purification purposes and is lyophilized for storage .
CD32a plays a significant role in the immune system by mediating various immune responses. It is involved in:
The study of CD32a is important for understanding its role in various diseases and therapeutic applications. For instance, CD32a is implicated in autoimmune diseases, where its interaction with immune complexes can contribute to inflammation and tissue damage. Additionally, targeting CD32a with therapeutic antibodies is being explored as a potential treatment for certain cancers and inflammatory conditions .
Recombinant CD32a expressed in HEK293 cells is widely used in research to: