FCGRT Mouse is a heterodimeric protein complex composed of two distinct subunits: the FCGRT (Fc Fragment of IgG Receptor and Transporter) and B2M (beta-2 microglobulin) . This protein belongs to the immunoglobulin superfamily and functions as a cell surface receptor with specialized binding properties .
The FCGRT/B2M complex is predominantly localized to the endosome membrane, where it functions to intercept and bind IgG molecules that have been internalized by cells . This strategic localization is critical for its function in IgG recycling and protection from degradation.
Available recombinant mouse FCGRT/B2M heterodimer proteins exhibit high purity levels, typically >95% as determined by SDS-PAGE and SEC-MALS (Size Exclusion Chromatography with Multi-Angle Light Scattering) . These proteins are often produced in mammalian expression systems such as HEK293 cells to ensure proper folding and post-translational modifications .
The mouse FCGRT protein performs several critical immunological functions that are essential for both neonatal immunity and lifelong IgG homeostasis.
The primary evolutionary function of mouse FCGRT is to transfer passive humoral immunity from mother to offspring . This process involves:
Binding of maternal IgG at the apical surface of the intestinal epithelium in neonates
Formation of FCGRT-IgG complexes
Transcytosis of these complexes across the intestinal epithelium
Release of IgG from FCGRT into the neonate's bloodstream or tissue fluids
This mechanism enables the transfer of protective antibodies to offspring before their own immune system becomes fully functional, providing critical early-life protection against pathogens .
Throughout life, mouse FCGRT contributes significantly to effective humoral immunity by:
Binding monomeric IgG in acidic endosomes of endothelial and hematopoietic cells
Recycling bound IgG to the cell surface
This pH-dependent binding mechanism (strong binding at acidic pH, weak binding at neutral pH) allows FCGRT to salvage IgG from lysosomal degradation, thereby extending the half-life of circulating IgG antibodies . This process is crucial for maintaining adequate levels of protective antibodies in the bloodstream.
Beyond its role in IgG maintenance, mouse FCGRT also regulates the homeostasis of serum albumin, the most abundant circulating protein . Similar to its interaction with IgG, FCGRT binds albumin in a pH-dependent manner and protects it from degradation, thereby contributing to the regulation of plasma oncotic pressure and the transport of various molecules in the bloodstream.
Various experimental models and research tools have been developed to study mouse FCGRT function and applications.
FCGRT knockout mice (B6.129X1-Fcgrt^tm1Dcr/DcrJ) provide valuable insights into the physiological roles of this protein . These genetically modified mice:
Are unable to recycle IgG or albumin
Degrade IgG at an accelerated rate
Display low plasma expression levels of both IgG and albumin
These phenotypic characteristics underscore the critical role of FCGRT in maintaining normal levels of these essential serum proteins.
Humanized FCGRT immune checkpoint knock-in mice represent advanced tools for therapeutic research . These models contain the human FCGRT gene instead of the mouse ortholog, enabling more translational studies of human therapeutic antibodies and their interactions with the human FcRn receptor in a living system.
A recent study has reported the development of a humanized ACE2, TMPRSS2, and FCGRT mouse model for studying SARS-CoV-2 infection and immune responses, providing direct evidence of the superior protective efficacy of hybrid immunity compared with vaccination alone . This model demonstrates the utility of FCGRT-modified mice in infectious disease research.
Several engineered cell lines have been developed for FCGRT research, including:
MDCK/Mouse FCGRT-P2A-mGFP&B2M Stable Cell Line: This cell line expresses mouse FCGRT fused with mGFP (modified Green Fluorescent Protein) and B2M, enabling visualization and cell-based binding assays .
The expression of these proteins can be verified through flow cytometry analysis using specific antibodies against mouse B2M or through direct detection of the GFP signal .
Various reagents are available for FCGRT research, including:
Recombinant Mouse FCGRT/B2M Heterodimer Proteins: These are available with different tags (His-tag, Avi-tag) and produced in various expression systems .
Anti-Mouse FCGRT/FCRN Polyclonal Antibodies: These can be used for applications such as ELISA, immunohistochemistry (IHC), and Western blotting (WB) .
Mouse FCGRT exhibits specific binding interactions with various ligands, which can be quantified using surface plasmon resonance (SPR) and bio-layer interferometry (BLI) assays.
Mouse FCGRT/B2M heterodimer protein demonstrates high-affinity binding to various antibodies:
Binding to Herceptin with an affinity constant of 2.52 nM (measured by SPR)
Binding to Herceptin with an affinity constant of 1.13 nM (measured by BLI)
Binding to Mouse IgG Fc with an affinity constant of 14.5 nM (measured by BLI)
These binding properties are critical for understanding the pharmacokinetics of therapeutic antibodies in mouse models.
Mouse FCGRT also binds to mouse serum albumin:
Binding to biotinylated mouse serum albumin with an affinity constant of 377 nM (SPR)
Binding to mouse serum albumin with an affinity constant of 0.376 μM (SPR)
These measurements confirm the dual role of FCGRT in maintaining both IgG and albumin homeostasis, though with differing affinities.
The understanding of mouse FCGRT has numerous research and therapeutic applications.
Knowledge of mouse FCGRT binding properties is essential for:
Designing therapeutic antibodies with improved half-life
Understanding species-specific differences in antibody pharmacokinetics
Interpreting preclinical data from mouse models
Researchers can modify the Fc region of antibodies to enhance or reduce binding to FCGRT, thereby controlling circulation time and tissue distribution .
The role of FCGRT in immune function makes it relevant to vaccine research:
FCGRT-mediated transport of antibodies across mucosal surfaces can influence vaccine efficacy
Understanding maternal antibody transfer through FCGRT is crucial for timing infant vaccinations
Humanized FCGRT mice provide valuable models for vaccine testing
The binding and transport capabilities of FCGRT can be exploited for drug delivery:
Fc-fusion proteins that leverage FCGRT binding for extended circulation
Albumin-conjugated drugs that utilize FCGRT-mediated recycling
Targeted delivery systems that exploit FCGRT expression in specific tissues
Research on mouse FCGRT continues to expand, with several promising directions:
Further characterization of tissue-specific FCGRT expression and function
Development of more sophisticated humanized models incorporating multiple human genes
Exploration of FCGRT polymorphisms and their impact on immune function
Investigation of FCGRT-targeting strategies for modulating autoimmune diseases
Despite significant progress, challenges remain in translating findings from mouse FCGRT studies to human applications due to species-specific differences in binding properties and expression patterns.
FCGRT mouse models are genetically modified mice with alterations to the gene encoding the neonatal Fc receptor (FcRn), a critical protein that binds to the Fc region of immunoglobulin G (IgG) antibodies. These models serve as essential tools for studying antibody trafficking, recycling mechanisms, and evaluating therapeutic antibodies.
The significance of these models stems from FcRn's crucial role in extending antibody half-life through a pH-dependent recycling process. FcRn binds to the Fc region of monomeric IgG in acidic endosomes, protecting antibodies from degradation and returning them to circulation, thereby significantly extending their lifespan in the body . This mechanism is fundamental for maintaining effective humoral immunity and has major implications for antibody-based therapeutics.
Most notably, humanized FCGRT mice have the mouse FCGRT gene replaced with the human version, creating a physiologically relevant system for evaluating human antibody dynamics. This overcomes the limitation that mouse FcRn has different binding characteristics with human IgG compared to human FcRn, making these models crucial for translational research .
The FcRn receptor facilitates several critical immunological processes through its specialized binding properties:
Maternal-fetal antibody transfer: FcRn enables the transfer of maternal IgG antibodies across the placenta to provide passive immunity to the developing fetus.
Neonatal immunity acquisition: In newborn animals, FcRn mediates the selective uptake of IgG from maternal milk. The process involves IgG binding at the apical surface of the intestinal epithelium, followed by transcytosis of the FcRn-IgG complexes across the intestinal epithelium, and release of IgG into the bloodstream or tissue fluids .
IgG recycling: Throughout life, FcRn contributes to effective humoral immunity by recycling IgG and extending its half-life in circulation. This occurs when monomeric IgG binds to FcRn in acidic endosomes of endothelial and hematopoietic cells, preventing lysosomal degradation. The complex is then recycled to the cell surface where IgG is released back into circulation at neutral pH .
Albumin homeostasis: Beyond antibody regulation, FcRn also regulates homeostasis of albumin, the most abundant circulating protein .
These mechanisms collectively explain why IgG has a significantly longer half-life (approximately 21 days in humans) compared to other proteins of similar size.
Several critical differences exist between humanized FCGRT and wild-type mouse models that significantly impact experimental outcomes:
| Feature | Wild-type Mouse | Humanized FCGRT Mouse |
|---|---|---|
| Binding affinity for human IgG | Unusually high affinity | Physiologically relevant affinity (lower) |
| Human antibody half-life | Artificially extended | More predictive of human pharmacokinetics |
| Anti-human IgG response | Develops immune response against human antibodies | Reduced immunogenicity against human antibodies |
| Competitive environment | Lacks human IgG competition | Can be engineered to have human IgG competition |
| Translational value | Limited for human therapeutic development | Higher predictive value for human applications |
Wild-type mice express mouse FcRn, which has a higher affinity for human IgG than human FcRn does . This results in artificially extended half-lives of human antibodies in wild-type mice, complicating pharmacokinetic predictions. Additionally, wild-type mice can mount an anti-human IgG response that rapidly clears antibodies via immune complex-mediated pathways .
In contrast, humanized FCGRT mice provide a more physiologically appropriate environment for testing human antibodies, with binding characteristics that better match human conditions. Some models, like the Tg32-hFc strain, even produce chimeric antibodies with human Fc regions that compete with administered antibodies, creating a more realistic competitive environment .
The presence of endogenous IgG in FCGRT humanized mice creates a competitive environment that significantly impacts experimental outcomes, particularly for therapeutic antibody evaluation:
Concentration-competition effect: In models like Tg32-hFc that produce chimeric IgG with humanized Fc regions, the endogenous production of these antibodies significantly dampens the serum half-life of administered humanized monoclonal antibodies through an FCGRT-dependent competitive mechanism . This competition can be further heightened by immunization, which increases endogenous antibody levels.
Physiological relevance: This competition effect actually provides a more physiologically appropriate environment for preclinical assessment of human IgG-based biologics, as it better mimics the human condition where therapeutic antibodies must compete with endogenous IgG for FcRn binding .
Dose adjustment considerations: Researchers must account for this competition when determining dosing regimens, as higher doses may be required to achieve target serum levels compared to non-competitive systems.
Experimental design implications: When using these models, it's essential to measure baseline endogenous IgG levels and consider how they might vary between individual animals. Including measurement of both total IgG and specific antibody concentrations can help interpret pharmacokinetic data more accurately.
The translational value of this competition is substantial—it provides a more realistic prediction of how therapeutic antibodies will behave in human patients where similar competition for FcRn binding occurs.
Several sophisticated methodological approaches are available for characterizing FCGRT function in transgenic mouse models:
Pharmacokinetic profiling: Tracking labeled antibodies over time to determine half-life, clearance rates, and volume of distribution. This should include multiple time points and appropriate controls with known FcRn binding properties.
In vivo competition studies: Administering differentially labeled antibodies with varying affinities for FcRn simultaneously to assess competitive binding effects under physiological conditions.
Tissue-specific expression analysis: Using immunohistochemistry, flow cytometry, or single-cell RNA sequencing to characterize FcRn expression patterns across different tissues and cell types.
pH-dependent binding assays: Employing surface plasmon resonance with pH gradients to measure the binding kinetics of FcRn-IgG interactions at different pH values that mimic various cellular compartments.
Integrative approaches using multi-humanized models: For specific applications like SARS-CoV-2 research, triple knock-in (TKI) mouse models expressing human ACE2, TMPRSS2, and FCGRT can provide comprehensive systems for evaluating human antibody therapeutics against infectious diseases .
ELISA-based quantification: Using enzyme-linked immunosorbent assays for precise detection of FcRn levels in mouse serum, plasma, and cell culture supernatants. Commercial kits are available with detection ranges of 0.312-20ng/ml and sensitivity of approximately 0.17ng/mL .
These methodologies provide researchers with robust tools to characterize FcRn biology in transgenic mouse models and evaluate therapeutic antibody candidates with greater translational relevance.
Optimizing experimental design for therapeutic antibody development using FCGRT humanized mice requires careful consideration of several factors:
Selection of appropriate model: Different FCGRT humanized mouse strains have distinct characteristics. For example, the Tg32-hFc strain produces chimeric antibodies with human Fc regions at physiologically relevant levels, providing a competitive environment that better mimics human conditions . Choose a model that aligns with your specific research question.
Control for competition effects: Account for the competition between endogenous and administered antibodies by including appropriate controls and considering dose adjustments. Measure baseline endogenous IgG levels before experiment initiation.
Dosing strategy optimization:
Implement multiple dose levels to characterize dose-dependent effects
Consider more frequent early sampling to capture distribution phase accurately
Adjust study duration based on expected half-life in humanized models (typically shorter than in wild-type mice)
| Study Parameter | Wild-type Mice | FCGRT Humanized Mice |
|---|---|---|
| Sampling schedule | Standard intervals | More frequent early timepoints |
| Study duration | Extended (due to artificially long half-life) | Shorter (more physiologically relevant) |
| Dose levels | Lower doses may appear effective | May require higher doses due to competition |
| Controls | Species-matched antibodies | Include FcRn-binding variants as controls |
Integration with disease models: For therapeutic applications, consider using disease models with humanized FCGRT to evaluate both pharmacokinetics and efficacy simultaneously.
Translation to humans: Develop scaling factors between humanized mice and humans based on parameters such as body weight, plasma volume, and FcRn expression levels to support human dose predictions.
By implementing these design considerations, researchers can generate more translatable data for therapeutic antibody development and reduce late-stage failures in clinical development.
FCGRT polymorphisms have significant implications for therapeutic antibody development that researchers must consider:
Variable binding kinetics: Polymorphisms in human FCGRT can alter binding affinity for IgG, affecting recycling efficiency and antibody half-life. This variation may explain differential responses to antibody therapeutics observed across patient populations.
Population considerations: Most humanized FCGRT mouse models contain a single FCGRT variant, which may not represent the diversity present in human populations. Results obtained from these models may not generalize equally across all human genetic backgrounds.
Therapeutic optimization challenges: Antibodies optimized for one FCGRT variant may perform differently with others, potentially affecting efficacy and dosing requirements. This presents challenges for therapeutic development and personalized medicine approaches.
Disease associations: Some FCGRT polymorphisms are associated with autoimmune or inflammatory conditions, which may influence baseline IgG levels and recycling dynamics. These associations should be considered when interpreting data for disease-specific applications.
Model development opportunities: There is a need for developing multiple humanized FCGRT mouse lines with different human polymorphic variants to better represent human genetic diversity. Isogenic lines differing only in FCGRT polymorphisms would be valuable for comparative studies.
Researchers developing therapeutic antibodies should consider characterizing their candidates across multiple FCGRT variants when possible and be aware of the limitations in extrapolating results across diverse human populations.
Translating findings from FCGRT mouse models to humans requires systematic approaches to address species-specific differences:
Multi-humanized models: For certain applications, such as SARS-CoV-2 research, utilize more comprehensively humanized models. The triple knock-in mouse model expressing human ACE2, TMPRSS2, and FCGRT provides a more integrated humanized system for testing human-specific therapeutics .
Cross-reactivity assessment: When possible, test therapeutic antibodies for binding to both human and mouse targets. Consider developing cross-reactive antibodies for mechanistic studies to minimize confounding factors related to target binding differences.
Fc receptor network considerations: Human IgG may interact differently with mouse Fcγ receptors compared to human receptors. For comprehensive evaluation, consider complementary studies in models with humanized Fcγ receptors or use Fc-engineered variants that minimize binding to non-FcRn receptors.
Comparative controls: Include species-matched controls in experimental design—mouse antibodies in wild-type mice and human antibodies in humanized mice—to establish baseline comparisons. Test multiple Fc variants with known differential binding to human FcRn as internal controls.
Bridging studies: Conduct parallel studies in humanized mice, non-human primates, and (when available) human samples to establish correlations between model systems and improve predictive power. Determine species-specific correction factors for pharmacokinetic parameters.
Physiological context awareness: Consider differences in immune system development, tissue expression patterns of FcRn, and baseline IgG levels when interpreting results. Some phenomena observed in mice may differ in humans due to these contextual factors.
By systematically addressing these considerations, researchers can maximize the translational value of humanized FCGRT mouse models while accounting for their inherent limitations.
FCGRT humanized mouse models have made significant contributions to SARS-CoV-2 research through specialized approaches:
Triple humanized models: Researchers have developed triple knock-in (TKI) mouse models expressing human ACE2, TMPRSS2, and FCGRT. These models support robust infection with both ancestral and emerging SARS-CoV-2 variants of concern while also allowing evaluation of human antibody therapeutics with physiologically relevant pharmacokinetics .
Evidence for hybrid immunity: Studies using these models have provided direct evidence supporting the superior protective efficacy of hybrid immunity (infection plus vaccination) compared to vaccination-only immunity against SARS-CoV-2 .
Convalescent plasma evaluation: The humanized FCGRT component allows for proper evaluation of human convalescent plasma as a potential therapeutic approach, with physiologically relevant antibody circulation times that better predict clinical outcomes.
Monoclonal antibody testing: These models permit more accurate assessment of therapeutic human monoclonal antibodies engineered for prolonged half-lives, offering a platform that better predicts clinical pharmacokinetics and efficacy.
Limitations to consider: While these models offer significant advantages, researchers should be aware of their limitations. As noted in the literature, "In humans, comorbidities such as obesity and compromised immune systems have been linked with worsened SARS-CoV-2 infection outcomes, but comorbidities were not modelled in this study" . Additionally, while these models focus on FcRn interactions, antibody interactions with other human Fc receptors could play additional roles in SARS-CoV-2 immunity.
These specialized mouse models continue to serve as valuable tools for evaluating both the fundamental biology of SARS-CoV-2 infection and potential therapeutic interventions.
Current FCGRT mouse models have several limitations that researchers are working to address:
Incomplete humanization: While FCGRT humanization addresses antibody recycling, most models retain mouse versions of other Fc receptors and immune components. This creates a chimeric system that may not fully recapitulate human antibody functions beyond half-life extension .
Tissue expression differences: Expression patterns of FcRn across tissues may differ between humanized mice and humans, potentially affecting tissue-specific antibody distribution and function. More detailed characterization of tissue-specific expression is needed.
Limited genetic diversity: Most current models represent a single FCGRT genetic variant, despite human populations exhibiting polymorphic variations that can affect antibody pharmacokinetics. Future models incorporating multiple human variants would better represent human diversity.
Integration challenges: When studying diseases like SARS-CoV-2, researchers need to combine FCGRT humanization with other humanized genes (ACE2, TMPRSS2) to create relevant models. While progress has been made with triple knock-in models , integration of additional human genes presents technical challenges.
Comorbidity modeling: Current models typically use healthy mice, whereas many human patients have comorbidities that affect immune function and drug disposition. As noted in recent research: "In humans, comorbidities such as obesity and compromised immune systems have been linked with worsened SARS-CoV-2 infection outcomes, but comorbidities were not modelled in this study" .
Future directions to address these limitations include:
Development of more comprehensively humanized immune systems
Creation of FCGRT polymorphism panels representing human genetic diversity
Integration of comorbidity factors into humanized models
Advanced tissue-specific humanization approaches using conditional expression systems
Development of computational approaches to bridge between model systems and humans
Addressing these limitations will enhance the predictive value of FCGRT mouse models for human therapeutic development.
Several complementary methods are recommended for accurate quantification of FCGRT expression in mouse models:
ELISA-based detection: Commercial ELISA kits specifically designed for mouse FCGRT/FcRn quantification offer high sensitivity (approximately 0.17ng/mL) and a detection range of 0.312-20ng/ml. These assays provide reliable quantification in mouse serum, plasma, and cell culture supernatants .
| Technical Specifications for FCGRT ELISA | Performance Characteristics |
|---|---|
| Detection Range | 0.312-20ng/ml |
| Sensitivity | 0.17ng/mL |
| Intra-assay CV | 5.9% |
| Inter-assay CV | 9.1% |
Immunohistochemistry: For tissue-specific expression patterns, immunohistochemistry using validated antibodies against the extracellular domain of FCGRT/FcRn can provide spatial information. Antibodies suitable for this application should be carefully selected based on validation for immunohistochemistry applications .
Flow cytometry: For cell-specific expression analysis, flow cytometry allows quantification of FCGRT at the single-cell level, enabling identification of expression heterogeneity across cell populations.
RT-qPCR: For transcriptional analysis, reverse transcription quantitative PCR offers a sensitive method to measure FCGRT mRNA levels across different tissues or under various experimental conditions.
Western blotting: For protein-level confirmation, western blotting using validated antibodies such as those targeting the extracellular domain can provide information about protein size and potential post-translational modifications .
Mass spectrometry: For absolute quantification, targeted mass spectrometry approaches using labeled peptide standards can provide highly accurate measurements of FCGRT protein levels.
When selecting methods, researchers should consider the specific research question, required sensitivity, and whether spatial or cellular resolution is needed. Combined approaches often provide the most comprehensive characterization of FCGRT expression.
Interpreting pharmacokinetic data from FCGRT humanized mouse models requires careful consideration of several key factors:
Competition effects: In models like Tg32-hFc that produce endogenous chimeric antibodies with human Fc regions, competition for FcRn binding significantly affects pharmacokinetics. The "endogenous production of the chimeric IgG1 significantly dampens serum half-life of administered humanized mAbs in an FCGRT-dependent manner consistent with an appreciable concentration-competition effect" . This competition must be accounted for when analyzing clearance rates.
Model-specific baseline: Each FCGRT humanized model has unique characteristics, so researchers should establish model-specific pharmacokinetic parameters rather than comparing directly to wild-type mice or other humanized strains without consideration of these differences.
Multi-compartment analysis: Simple elimination half-life calculations may be insufficient. Consider using multi-compartment modeling that can distinguish between distribution and elimination phases, particularly for novel antibody formats.
Comparative standards: Include well-characterized standard antibodies with known human pharmacokinetics as internal controls to enable relative comparisons.
Translational scaling: Develop allometric or physiologically-based pharmacokinetic models that incorporate species-specific differences in body weight, plasma volume, and FcRn expression levels to extrapolate to human predictions.
By considering these factors, researchers can more accurately interpret pharmacokinetic data from FCGRT humanized mice and make more reliable predictions for human applications, avoiding both over- and under-estimation of therapeutic antibody half-lives.
Selecting the optimal FCGRT mouse model requires careful alignment between model characteristics and specific research objectives:
Research purpose determination: First establish whether your primary goal is mechanistic understanding of FcRn biology or translational evaluation of therapeutic antibodies, as this fundamentally guides model selection.
Expression level considerations: Different models express human FCGRT at varying levels. Consider whether physiological expression levels are critical for your research question or if higher expression might be beneficial for certain applications.
Genetic background impacts: The background strain of FCGRT humanized mice can influence experimental outcomes through strain-specific immune characteristics. Select models with genetic backgrounds relevant to your disease model or research question.
Multi-humanized requirements: For infectious disease studies (like SARS-CoV-2) or complex immune evaluations, consider triple knock-in models expressing multiple humanized genes (ACE2, TMPRSS2, FCGRT) for a more comprehensive system .
Practical considerations: Technical factors including breeding efficiency, model availability, cost, and existing validated protocols may also influence selection decisions.
| Research Focus | Recommended Model Characteristics |
|---|---|
| Therapeutic antibody PK/PD | Models with endogenous human-compatible IgG competition |
| Basic FcRn biology | Models with minimal confounding factors |
| Infectious disease therapeutics | Multi-humanized models (e.g., TKI with ACE2/TMPRSS2/FCGRT) |
| Fc engineering optimization | Models with human FCGRT and minimal competition |
The Fc fragment of IgG receptor and transporter, also known as the neonatal Fc receptor (FcRn), plays a crucial role in the immune system by mediating the transport and recycling of immunoglobulin G (IgG) antibodies. This receptor is essential for maintaining the long half-life of IgG and for transferring maternal antibodies to the fetus, providing passive immunity.
FcRn is a heterodimer composed of two subunits: the alpha chain (FCGRT) and beta-2-microglobulin (B2M). The alpha chain binds to the Fc region of IgG, while B2M stabilizes the structure. This receptor is structurally similar to major histocompatibility complex (MHC) class I molecules .
The primary function of FcRn is to protect IgG from lysosomal degradation by binding to it in acidic environments (such as endosomes) and releasing it at neutral pH (such as in the bloodstream). This recycling process extends the half-life of IgG antibodies, allowing them to persist longer in the circulation .
During pregnancy, FcRn is responsible for transporting IgG antibodies from the mother to the fetus across the placenta. This transfer provides the fetus with passive immunity, protecting it from infections during the early stages of life. The interaction between FcRn and IgG is pH-dependent, with binding occurring at acidic pH and release at neutral pH .
FcRn has become a target for therapeutic interventions, especially in autoimmune diseases. By modulating FcRn activity, it is possible to alter the half-life of pathogenic IgG antibodies, reducing their levels in the circulation. This approach has shown promise in treating conditions such as immune thrombocytopenia, myasthenia gravis, and systemic lupus erythematosus .