FCGR2B exists as two splice variants (FCGR2B1 and FCGR2B2) generated via mRNA splicing :
Isoform | Exon C1 | Cellular Localization | Function |
---|---|---|---|
FCGR2B1 | Included | B-cell membrane | Long cell-surface retention; inhibits B-cell activation |
FCGR2B2 | Excluded | Myeloid cell cytoplasm | Rapid internalization; modulates phagocytosis |
Both isoforms share a 95% extracellular identity with activating FcγRs (e.g., FCGR2A) but feature a unique immunoreceptor tyrosine-based inhibitory motif (ITIM) in their cytoplasmic domain .
Attribute | Detail |
---|---|
Gene Location | Chromosome 1q23.3 |
Protein Length | 310 amino acids (isoform B1) |
Domains | Extracellular IgG-binding domain, transmembrane helix, ITIM motif |
Expression | B cells, myeloid dendritic cells, basophils, liver endothelial cells |
FCGR2B binds immune complexes via its extracellular domain and recruits phosphatases (e.g., SHIP1/2) through its ITIM motif to suppress activating signals .
FCGR2B modulates immune responses through:
B-Cell Regulation: Increases the activation threshold of B-cell receptors (BCRs), limiting antibody production and promoting apoptosis .
Myeloid Cell Inhibition: Counterbalances activating FcγRs on dendritic cells and macrophages, reducing pro-inflammatory cytokine release .
Germinal Center Control: Facilitates antigen retention on follicular dendritic cells (FDCs), ensuring high-affinity antibody selection .
ITIM-Dependent Signaling: Phosphorylated ITIM recruits SHIP1/2, inhibiting Ras/MAPK and PLCγ/PKC pathways .
Lipid Raft Exclusion: The T232 transmembrane polymorphism disrupts lipid raft localization, impairing inhibitory function .
FCGR2B dysfunction is linked to systemic lupus erythematosus (SLE) and rheumatoid arthritis:
The FCGR2B-232T variant (rs1050501) reduces inhibitory capacity, increasing SLE risk (OR = 1.8–2.5 in Asians and Europeans) .
Impaired FcγRIIB expression on memory B cells correlates with SLE severity .
The FCGR2B-232T allele confers protection against severe malaria (OR = 0.56 in East Africans) .
FCGR2B internalizes immune complexes in liver endothelial cells, limiting systemic inflammation .
FCGR2B enhances anti-tumor responses by cross-linking checkpoint antibodies (e.g., anti-CD137) .
Tumor-infiltrating CD8+ T cells upregulate FCGR2B, which suppresses anti-melanoma immunity .
Model/Tool | Application | Source |
---|---|---|
FCGR2B CHO-K1 cell line | Ligand binding assays | BPS Bioscience |
Humanized NSG mice | Autoimmunity studies | PNAS |
AlphaFold-predicted structures | Protein interaction mapping | Protein Atlas |
FCGR2B (Fc fragment of IgG receptor IIb) is a low-affinity inhibitory receptor for the Fc region of immunoglobulin gamma (IgG). It is the only inhibitory type I FcγR in humans and mice . Its primary functions include:
Participation in the phagocytosis of immune complexes
Regulation of antibody production by B lymphocytes
Inhibition of activating FcγR functions, including pro-inflammatory cytokine release
Downregulation of B cell receptor signaling
Methodologically, researchers investigating FCGR2B function should employ cellular assays that measure B cell activation, proliferation, and antibody production in the presence of FCGR2B crosslinking. Additionally, phagocytosis assays using immune complexes can assess the regulatory role of FCGR2B in myeloid cells.
FCGR2B exists in two major isoforms (FCGR2B1 and FCGR2B2) created through alternative mRNA splicing . The key structural characteristics include:
An Immunoreceptor Tyrosine-based Inhibitory Motif (ITIM) in the cytoplasmic region
The FCGR2B1 isoform includes the C1 exon sequence, resulting in membrane tethering on B cells
The FCGR2B2 isoform excludes the C1 exon sequence, allowing faster internalization in myeloid cells
Extracellular domains that are 95% identical to FCGR2A and almost completely identical to FCGR2C
Canonical protein length of 310 amino acids with a molecular mass of approximately 34 kDa
The distinguishing feature of FCGR2B is its ITIM motif, which contrasts with the Immunoreceptor Tyrosine-based Activation Motifs (ITAMs) found in activating FcγRs. Researchers should use isoform-specific primers in RT-PCR assays and domain-specific antibodies to differentiate FCGR2B from other family members in experimental studies.
FCGR2B expression varies across immune cell populations and is subject to complex regulation:
FCGR2B1 is highly expressed by B cells, with lower levels on monocytes
FCGR2B2 is highly expressed on basophils and at low levels on monocytes
FCGR2B is co-expressed with activating FCGRA on circulating myeloid dendritic cells
Expression is positively regulated by IL-10 and IL-6, and negatively regulated by TNF-α, C5a, and IFN-γ
At least 10 single-nucleotide polymorphisms have been identified in the FCGR2B promoter region, with certain haplotypes leading to increased expression under both constitutive and stimulated conditions . To study expression regulation, researchers should employ quantitative PCR, flow cytometry, and cytokine stimulation assays with cell type-specific markers to accurately characterize expression patterns across different immune populations.
When FCGR2B is engaged, it initiates inhibitory signaling cascades that counteract activating signals:
The ITIM motif becomes phosphorylated, creating a docking site for phosphatases
Inositol phosphatases SHIP1 and SHIP2 are recruited to the phosphorylated ITIM
These phosphatases inhibit Ras activation and downregulate MAPK activity
PLCγ function is reduced, leading to decreased activation of PKC
Calcium mobilization is suppressed, inhibiting cellular activation
To study these signaling events, researchers should employ phospho-specific Western blotting, immunoprecipitation assays to detect protein-protein interactions, calcium flux measurements, and phosphoproteomic approaches to comprehensively map the signaling network. Genetic approaches using SHIP1/2 knockouts or inhibitors can help delineate the specific roles of these phosphatases in FCGR2B signaling.
FCGR2B plays a significant role in shaping the tumor microenvironment and contributing to immunosuppression:
FCGR2B may influence tumor occurrence, development, and invasion by modulating the immunosuppressive tumor microenvironment
In glioma, FCGR2B expression has been analyzed using databases such as TCGA, CGGA, and GEO, revealing correlations with immune scores and tumor grade
The receptor can inhibit anti-tumor immune responses by dampening antibody-dependent cellular cytotoxicity and phagocytosis
To investigate these mechanisms, researchers should:
Analyze FCGR2B expression in tumor tissue using immunohistochemistry and flow cytometry
Correlate expression with immune cell infiltration patterns using multiplex immunofluorescence or mass cytometry
Employ FCGR2B knockout or blocking approaches in tumor models to assess impact on tumor growth and anti-tumor immunity
Analyze the relationship between FCGR2B expression and response to immunotherapy
FCGR2B promoter polymorphisms significantly impact gene expression and disease susceptibility:
Ten single-nucleotide polymorphisms have been identified in the FCGR2B promoter region
Two functionally distinct haplotypes in the proximal promoter have been characterized
The less frequent promoter haplotype leads to increased reporter gene expression in both B lymphoid and myeloid cell lines
This haplotype shows significant association with systemic lupus erythematosus (SLE) (odds ratio = 1.65; p = 0.0054)
The association persists after adjustment for FCGR2A and FCGR3A polymorphisms (odds ratio = 1.72; p = 0.0083)
Methodologically, researchers should employ:
Luciferase reporter assays to assess promoter variant function
EMSA (electrophoretic mobility shift assay) to identify differential transcription factor binding
Case-control association studies with appropriate population stratification controls
Functional validation in primary cells from genotyped individuals
Haplotype analysis to account for linkage disequilibrium across the FCGR locus
FCGR2B expression has emerging value as a prognostic biomarker in various cancers:
In glioma, researchers have used survival receiver operating characteristic curves, univariate and multivariate Cox analysis to evaluate FCGR2B as a prognostic marker
Nomograms incorporating FCGR2B expression with clinicopathological features have been constructed to predict 1-year, 2-year, and 3-year survival
Calibration curves have been used to evaluate the accuracy of survival predictions
To develop and validate FCGR2B as a prognostic biomarker, researchers should:
Analyze large, well-annotated patient cohorts with comprehensive follow-up data
Employ multivariate models that account for established prognostic factors
Validate findings across independent datasets using the same analytical methods
Consider cell type-specific FCGR2B expression using single-cell approaches
Correlate expression with response to specific therapies, particularly immunotherapies
The significance of FCGR2B expression may vary across cancer types, necessitating tissue-specific validation and potentially different cutoff values for different malignancies.
FCGR2B functions within a complex network of immune regulatory mechanisms:
Correlation analyses between FCGR2B expression and various immune checkpoints have been performed using CGGA and TCGA datasets
The relationship between FCGR2B expression and tumor mutation burden (TMB) has been investigated in glioma
These interactions provide context for understanding FCGR2B's role in immune regulation
To study these relationships, researchers should:
Perform co-expression analyses of FCGR2B with established immune checkpoints (PD-1, CTLA-4, LAG-3)
Investigate the impact of combined blockade of FCGR2B and other checkpoints in functional assays
Analyze TMB in relation to FCGR2B expression across cancer types using whole-exome sequencing data
Employ multiparameter flow cytometry or mass cytometry to analyze co-expression patterns at the single-cell level
Develop network models incorporating FCGR2B with other immune regulatory molecules
Effective analysis of FCGR2B expression in clinical samples requires consideration of multiple technical factors:
Transcriptomic analysis: RNA sequencing or microarray analysis using databases like TCGA, CGGA, and GEO
Protein detection: Immunohistochemistry, flow cytometry, or Western blotting with validated antibodies
Single-cell approaches: scRNA-seq or mass cytometry for cell type-specific expression patterns
Researchers should consider:
Appropriate controls and normalization strategies to account for sample heterogeneity
Antibody specificity given the high homology with other FCGR family members
Distinction between FCGR2B1 and FCGR2B2 isoforms using specific primers
Post-translational modifications that may affect protein detection
Correlation with clinical parameters using appropriate statistical methods
For quantitative analysis, it's essential to establish standardized protocols that can be reproduced across different laboratories, particularly for potential diagnostic applications.
Evaluating the functional impact of FCGR2B genetic variants requires multiple complementary approaches:
Luciferase reporter assays to assess promoter variant effects on gene expression
CRISPR/Cas9 gene editing to introduce specific variants in relevant cell types
Patient-derived cells with different variants can be compared in functional assays
In silico prediction tools to assess coding variant impact on protein structure
Specific functional readouts should include:
Expression level quantification (mRNA and protein)
Surface localization and receptor trafficking
IgG binding affinity measurements using surface plasmon resonance
ITIM phosphorylation and downstream signaling pathway activation
Inhibitory capacity in relevant cellular contexts (B cell activation, phagocytosis)
Case-control association studies remain essential for linking variants to disease phenotypes, as demonstrated in studies of FCGR2B promoter haplotypes and SLE .
When designing studies to investigate FCGR2B's role in disease pathogenesis, researchers should consider:
Cross-sectional and longitudinal approaches to capture disease dynamics
Integration of genetic, transcriptomic, and functional analyses
Appropriate disease and healthy control populations with demographic matching
Animal models with relevant FCGR2B modifications
A comprehensive study design might include:
Genetic association analysis using well-characterized patient cohorts
Expression analysis in affected tissues using multiple methodologies
Functional assays with patient-derived cells
Animal models to validate mechanisms in vivo
Therapeutic targeting approaches to assess the impact of FCGR2B modulation
For autoimmune diseases like SLE, where FCGR2B promoter variants have been implicated , longitudinal studies during disease flares and remissions can provide insights into how FCGR2B contributes to disease dynamics.
Analyzing FCGR2B in large genomic datasets requires specialized bioinformatic approaches:
Differential expression analysis between high and low FCGR2B expression groups
GO, KEGG, and GSEA enrichment analyses to identify associated biological processes and pathways
Co-expression network analysis to identify genes functionally related to FCGR2B
Recommended analytical tools include:
"Limma" and "pheatmap" packages for differential gene expression analysis and visualization
"ComplexHeatmap", "limma", and "ggpubr" packages for correlation analyses
ESTIMATE algorithm to calculate immune and stromal scores in tumor samples
"timeROC" for receiver operating characteristic curve analysis in survival studies
Researchers should prioritize:
Standardized data preprocessing and normalization
Robust statistical methods with appropriate multiple testing correction
Validation in independent datasets
Integration of multiple data types when available
Consideration of confounding factors such as batch effects and platform differences
Conflicting data on FCGR2B expression is common and requires careful interpretation:
Technical factors: Different methodologies may yield varying results
Sample heterogeneity: Differences in patient populations, disease stages, and cellular composition
Isoform-specific detection: Studies may differentially detect FCGR2B1 versus FCGR2B2
Reference gene selection: Different normalization strategies affect relative expression values
To reconcile conflicting findings, researchers should:
Critically evaluate methodological details, including primer/antibody specificity
Consider cellular source of expression in heterogeneous samples
Assess whether differences reflect true biological variation
Perform meta-analyses when sufficient comparable studies exist
Design validation studies that specifically address discrepancies
When analyzing public database data (TCGA, CGGA, GEO), awareness of batch effects, platform differences, and potential confounding factors is essential .
Robust statistical methodologies are essential for analyzing FCGR2B's relationship with clinical outcomes:
Survival analysis: Kaplan-Meier methods with log-rank tests to compare high versus low FCGR2B expression groups
Multivariate Cox regression models to adjust for confounding clinical variables
Time-dependent ROC curves to evaluate prognostic value at different timepoints
Nomogram construction to develop predictive models incorporating FCGR2B
Key statistical considerations include:
Sample size calculation to ensure adequate power
Multiple testing correction to control false discovery rate
Assessment of effect sizes with confidence intervals
Validation in independent cohorts
For genetic association studies, adjustment for linkage disequilibrium with other FCGR genes is crucial, as demonstrated in studies of FCGR2B promoter haplotypes in SLE .
Distinguishing direct FCGR2B effects from indirect consequences on the immune microenvironment requires:
Cell type-specific analyses: Single-cell approaches to identify the source and targets of FCGR2B action
Deconvolution methods: Computational approaches to estimate cell type proportions in bulk tissue samples
Conditional knockout models: Cell type-specific FCGR2B deletion to isolate direct effects
In vitro co-culture systems: Controlled experimental settings to study cellular interactions
Analytical approaches include:
Correlation analysis between FCGR2B expression and immune cell infiltration patterns
Assessment of immune and stromal scores in relation to FCGR2B expression
Pathway analysis to identify mechanisms connecting FCGR2B to broader immune changes
Integration of spatial information through techniques like spatial transcriptomics
These approaches are particularly relevant in tumor contexts, where FCGR2B may influence the immunosuppressive microenvironment .
Developing therapeutic strategies targeting FCGR2B requires careful consideration of:
Cell type specificity: Targeting FCGR2B on specific cell populations to avoid unintended effects
Genetic variation: Accounting for FCGR2B polymorphisms that may affect therapeutic response
Pathway redundancy: Addressing compensatory mechanisms that may limit efficacy
Safety concerns: Mitigating risks of excessive immune activation or suppression
Key development strategies include:
Antibody engineering to modulate FCGR2B function or bypass its inhibitory effects
Cell-specific delivery approaches for genetic interventions
Combination approaches targeting complementary pathways
Biomarker development to identify patients most likely to benefit
In oncology, FCGR2B targeting might enhance efficacy of antibody therapies by preventing inhibitory signaling, while in autoimmunity, FCGR2B agonism could potentially suppress pathogenic immune responses. Both approaches require careful preclinical validation before clinical translation.
CD32, also known as Fc gamma receptor II (FcγRII), is a family of cell membrane receptor proteins that play a crucial role in the immune system. These receptors are part of the immunoglobulin superfamily and are primarily involved in the regulation of immune responses. CD32 receptors are encoded by the mRNA splice variants of three highly related genes: FCGR2A, FCGR2B, and FCGR2C .
CD32 receptors are primarily found on the surface of leukocytes, including macrophages, neutrophils, and some subsets of T cells. They are responsible for binding the Fc region of immunoglobulin G (IgG) antibodies. This binding triggers various immune responses, including phagocytosis, antibody-dependent cellular cytotoxicity (ADCC), and the release of inflammatory mediators .
The CD32 family consists of three main isoforms:
Recombinant CD32 proteins are produced using recombinant DNA technology, which involves inserting the gene encoding CD32 into a suitable expression system, such as bacteria, yeast, or mammalian cells. This allows for the large-scale production of CD32 proteins with high purity and consistency .
Recombinant CD32 proteins are used in various research and clinical applications, including:
CD32 receptors play a critical role in the regulation of immune responses and are implicated in various diseases. Dysregulation of CD32 signaling has been associated with autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). In these conditions, the balance between activatory and inhibitory CD32 isoforms is disrupted, leading to excessive immune activation and tissue damage .
CD32 receptors are also involved in the immune response to infections and cancer. For example, FcγRIIA (CD32a) is known to enhance the phagocytosis of opsonized pathogens, while FcγRIIB (CD32b) helps to prevent excessive inflammation during chronic infections .