IGHG3 Antibody

Shipped with Ice Packs
In Stock

Description

Genetic Diversity and Population-Specific Variations

The IGHG3 gene exhibits significant polymorphism across populations:

VariantFunctional ImpactPopulation Frequency
Hinge length (3 exons)Reduced phagocytosis/neutralization capacityHigher in African descent
IGHG3*04 (2-exon hinge)Enhanced ADCC activityRare (1% in studied cohorts)
435H (rs201919874)Prolonged half-life via FcRn bindingHigher in East Asians and Africans
291L/292W/296F allotypesReduced ADCCHigher in Europeans

Functional Roles in Immune Responses

IGHG3 antibodies mediate diverse effector functions:

  • Antiviral Activity:

    • Longer hinges enhance neutralization of SARS-CoV-2 by improving epitope accessibility .

    • Shorter hinges correlate with critical COVID-19 (OR = 2.79, 95% CI = 1.66–4.65) .

  • FcγR Interactions:

    • Allotypes with phenylalanine (296F) or tryptophan (292W) reduce FcγRIIIa affinity and ADCC .

    • Short-hinge variants (e.g., IGHG3*04) increase ADCC against HIV and cancer cells .

Infectious Diseases

  • COVID-19: Low IgG3 titers and short-hinge alleles predict severe outcomes .

  • HIV: Longer hinges improve phagocytosis but reduce ADCC .

Autoimmune Diseases

Systemic Lupus Erythematosus (SLE):

BiomarkerLevel in SLE vs HCCorrelation with Disease Activity
Serum IGHG3478.1 ± 160.9 vs 364.4 μg/mLAnti-dsDNA antibodies (r = 0.22, p = 0.003)
Urinary IGHG364.0 ± 74.5 vs 27.1 ng/mLSLE Disease Activity Index (r = 0.332, p < 0.01)
Salivary IGHG33,078.9 ± 2,473.8 vs 1,413.6 ng/mLESR (r = 0.173, p = 0.024)

Urinary IGHG3 levels are elevated in lupus nephritis (119.55 ± 110.0 vs 49.8 ± 54.4 ng/mL, p < 0.01) .

Therapeutic and Diagnostic Implications

  • Diagnostics: Urinary IGHG3 shows high diagnostic accuracy for SLE (AUC = 0.816 for nephritis) .

  • Therapeutic Design: Hinge-length engineering may optimize IgG3-based therapies for viral infections .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery times.
Synonyms
IGHG3Immunoglobulin heavy constant gamma 3 antibody; HDC antibody; Heavy chain disease protein antibody; Ig gamma-3 chain C region antibody
Target Names
IGHG3
Uniprot No.

Target Background

Function
The IGHG3 antibody targets the constant region of immunoglobulin heavy chains. Immunoglobulins, also known as antibodies, are membrane-bound or secreted glycoproteins produced by B lymphocytes. During the recognition phase of humoral immunity, membrane-bound immunoglobulins serve as receptors. Upon binding to a specific antigen, these receptors trigger the clonal expansion and differentiation of B lymphocytes into immunoglobulin-secreting plasma cells. Secreted immunoglobulins then mediate the effector phase of humoral immunity, leading to the elimination of bound antigens. The antigen binding site is formed by the variable domain of one heavy chain, in conjunction with its associated light chain. Therefore, each immunoglobulin possesses two antigen binding sites with remarkable affinity for a particular antigen. The variable domains are assembled through a process called V-(D)-J rearrangement and can undergo somatic hypermutations. These mutations, after exposure to antigen and selection, allow for affinity maturation towards a specific antigen.
Database Links

HGNC: 5527

OMIM: 147120

UniGene: Hs.510635

Subcellular Location
Secreted. Cell membrane.

Q&A

What is IGHG3 and how does it relate to the IgG3 antibody?

IGHG3 (immunoglobulin heavy constant gamma 3) is the gene that encodes the constant region of the heavy chain for IgG3 antibodies. IgG3 is the third most abundant of the four human IgG subclasses (IgG1, IgG2, IgG3, and IgG4), named in order of their relative serum concentrations. IgG3 is comprised of two identical heavy chains encoded by IGHG3, and two light chains (either Igκ or Igλ), linked by disulfide bonds . The gene is located on chromosome 14q32.33 and is involved in processes including antibacterial humoral response and complement activation through the classical pathway .

What are the unique structural features of IgG3 compared to other IgG subclasses?

IgG3 has several distinctive structural features:

  • Extended hinge region: The most notable feature of IgG3 is its uniquely extended hinge region, which can vary from 32 to 62 amino acids depending on the number of exon repeats (compared to shorter hinges in other IgG subclasses) .

  • Hinge composition: The hinge region is encoded by one A exon and from one to three 15 amino acid long B exons, depending on the G3m alleles .

  • Allotypic variations: IgG3 exhibits substantial polymorphism with variations in the hinge region length and amino acid composition, which affects its functional properties .

  • Flexibility: The elongated hinge provides IgG3 with greater flexibility, allowing for enhanced interaction with immune effector molecules .

This unique structure contributes to IgG3's superior capacity to mediate various effector functions, including intracellular antiviral immunity and complement activation .

What functional advantages does IgG3 demonstrate in antiviral immunity?

Research indicates that IgG3 possesses superior capacity to mediate intracellular anti-viral immunity compared with other IgG subclasses. This enhanced functionality depends primarily on:

  • TRIM21 pathway activation: IgG3 is the most potent activator of the cytosolic Fc receptor and ubiquitin ligase TRIM21, which plays a crucial role in intracellular antibody-mediated virus neutralization .

  • Complement activation: IgG3 demonstrates superior ability to activate the complement system, particularly through the C1/C4 pathway, enhancing viral neutralization .

  • Hinge-dependent enhancement: The extended and flexible hinge region of IgG3 allows for favorable activation of TRIM21 and complement components. Experiments have shown that IgG3 neutralized adenovirus (Ad5-mCherry) more efficiently than other IgG subclasses and induced over 2-fold more potent NF-κB responses in cellular infection assays .

  • Early response role: Several studies support that IgG3 is an anti-viral subclass that is produced acutely and acts before an IgG1 response becomes dominant during infection .

These characteristics make IgG3 particularly important in protection against viral pathogens, especially during early stages of infection.

How does IgG3 hinge length variation affect its functional properties?

The length and flexibility of the IgG3 hinge significantly impacts its functional properties:

  • TRIM21-dependent neutralization: Variants with more flexible hinges (such as the C1-3S variant with the first three cysteines replaced by serine) showed enhanced TRIM21-dependent neutralization and NF-κB induction activity beyond that of wild-type IgG3 .

  • Complement activation: Studies have shown that IgG3 with its natural extended hinge was more potent in activating complement than other subclasses. When the IgG3 hinge was swapped into IgG1, the resulting molecule gained enhanced C1/C4-dependent neutralization capacity .

  • ADCC activity: IgG3 antibodies with a shorter hinge (e.g., IGHG3*04 with 2 exons) exhibited stronger antibody-dependent cellular cytotoxicity (ADCC) capacity. This reduced hinge length was associated with increased ADCC against HIV-infected cell lines and CD20+ tumor cells .

  • Clinical implications: Hinge length variation has been associated with differential outcomes in various diseases. For example, shorter hinge length has been linked to increased inflammation and death in cerebral malaria .

These findings highlight the critical role of the IgG3 hinge region in determining its functional properties and suggest potential applications in antibody engineering for therapeutic purposes.

What techniques are used to determine IGHG3 hinge length polymorphisms?

Several methods have been developed to analyze IGHG3 hinge length polymorphisms:

These techniques enable researchers to characterize the polymorphic nature of IGHG3 and study its functional implications.

What are the recommended methods for analyzing IgG3 effector functions in experimental settings?

To analyze IgG3 effector functions in experimental settings, researchers can employ several methodologies:

  • TRIM21-dependent neutralization assay:

    • Infect wild-type and TRIM21 knockout cells (e.g., 293T cells) with a virus (e.g., Ad5-mCherry) in complex with titrated amounts of IgG3 antibodies

    • Determine infection levels relative to virus alone using flow cytometry

    • Calculate TRIM21-dependent neutralization as fold change between infection levels in wild-type and TRIM21 knockout cells

  • NF-κB reporter assay:

    • Use NF-κB reporter wild-type and TRIM21 knockout cells

    • Infect with virus complexed with IgG3 antibodies

    • Measure NF-κB activation to assess TRIM21-induced signaling

  • Complement activation assays:

    • C1/C4-dependent neutralization: Incubate virus with antibodies and complement-containing serum, then measure infectivity in cells lacking typical IgG Fc receptors

    • C4 convertase assay: Pre-incubate IgG3 with virus before adding human serum, then measure C4 cleavage over time using western blot

  • Antibody binding analysis:

    • ELISA: Measure binding of equimolar amounts of different IgG subclasses to virus particles

    • Surface plasmon resonance: Determine binding kinetics and affinity of IgG3 to different Fc receptors

  • ADCC assays:

    • Co-culture target cells expressing the antigen of interest with effector cells (e.g., NK cells) in the presence of IgG3 antibodies

    • Measure target cell killing through release of cytotoxic granules or cell death markers

These methods allow for comprehensive characterization of IgG3's unique effector functions and comparison with other IgG subclasses.

What are the key challenges in developing IgG3-based therapeutic antibodies?

Despite the functional advantages of IgG3, several challenges exist in developing IgG3-based therapeutics:

  • Physical and conformational stability: IgG3 demonstrates reduced physical and conformational stability compared to IgG1. Experimental analysis of anti-IL-8 IgG3 showed lower domain unfolding temperatures compared to its IgG1 counterpart, suggesting reduced shelf-life stability .

  • Solution viscosity: High-concentration formulations of IgG3 exhibit elevated solution viscosity compared to IgG1, which impacts manufacturability and injectability. This property is attributed to stronger protein-protein interactions in IgG3 .

  • Half-life considerations: Traditional IgG3 has a shorter serum half-life (approximately 7 days) compared to other IgG subclasses (21 days), although some allotypes with histidine at position 435 (435H) show prolonged half-life due to higher affinity to FcRn at low pH .

  • Allotypic variations: The high degree of polymorphism in IgG3 introduces complexity in predicting functional properties and immunogenicity across different populations .

  • Post-translational modifications: IgG3 may require more extensive monitoring of post-translational modifications for batch-to-batch consistency and shelf-life stability .

Addressing these challenges requires specialized approaches to antibody engineering and formulation development for IgG3-based therapeutics.

What biophysical parameters should be evaluated when developing IgG3 antibodies for therapeutic applications?

When developing IgG3 antibodies for therapeutic applications, several critical biophysical parameters should be evaluated:

  • Conformational stability:

    • Thermal unfolding transitions measured by differential scanning calorimetry (DSC) or nanodifferential scanning fluorimetry (nano-DSF)

    • Chemical denaturation studies to assess free energy of unfolding

  • Colloidal stability:

    • Second virial coefficient (B22) measurements

    • Dynamic light scattering (DLS) to determine diffusion coefficients and hydrodynamic radius

    • Zeta potential measurements to assess surface charge distribution

  • Solution viscosity:

    • Concentration-dependent viscosity profiles

    • Intrinsic viscosity and the Huggins coefficient determination

    • Assessment of protein-protein interactions that elevate solution viscosity in high-concentration formulations

  • Surface properties:

    • Hydrophobic interaction chromatography (HIC) to assess surface hydrophobicity

    • Charge variant analysis using capillary isoelectric focusing (cIEF)

    • Patch analysis to identify hydrophobic and charged patches on the protein surface

  • Molecular descriptors:

    • Computational analysis using homology modeling

    • Prediction of physicochemical properties including hydrophobic imbalance and buried surface area

    • Identification of charge patches and their potential impact on protein-protein interactions

ParameterExperimental TechniqueSignificance
Thermal stabilityNano-DSF/DSCPredicts shelf-life stability
Colloidal stabilityDLS, B22 measurementsIndicates aggregation propensity
Surface chargecIEFDetermines electrostatic interactions
ViscosityRheology measurementsImpacts manufacturability and delivery
Surface hydrophobicityHICPredicts non-specific interactions

These comprehensive evaluations help identify potential developability issues and guide rational engineering approaches to improve IgG3 antibodies for therapeutic applications.

How can the unique properties of IgG3 be leveraged for next-generation therapeutic antibodies?

The distinctive properties of IgG3 offer several opportunities for developing advanced therapeutic antibodies:

  • Enhanced intracellular immunity: IgG3's superior ability to activate TRIM21-mediated intracellular immunity could be exploited for developing antibodies against intracellular pathogens. The hinge-dependent enhancement of this pathway could be incorporated into antibody design through hinge engineering .

  • Complement activation optimization: The potent complement-activating properties of IgG3, particularly through the C1/C4 pathway, could be harnessed for cancer immunotherapy where complement-dependent cytotoxicity is desired. Specifically, the 17mer hinge region of IgG3 that enhances C1/C4-dependent neutralization could be incorporated into therapeutic antibodies .

  • Hinge engineering strategies:

    • Creating flexibility-optimized antibodies by replacing specific cysteines with serines (e.g., C1-3S variants) to enhance TRIM21 activity

    • Developing shorter hinge variants to improve ADCC against specific targets like CD20+ tumor cells

    • Creating IgG1/IgG3 hinge hybrids to combine the favorable pharmacokinetic properties of IgG1 with the enhanced effector functions of IgG3

  • Allotype selection: Selecting specific IgG3 allotypes (e.g., those with histidine at position 435) that demonstrate extended half-life comparable to other IgG subclasses while retaining enhanced effector functions .

  • Hexamerization technology: Leveraging IgG3's natural propensity for Fc:Fc interactions to develop antibodies with enhanced complement activation through controlled hexamerization, potentially applying mutations like E345R+E430G+S440Y that allow hexamerization in solution .

These approaches could yield novel therapeutic antibodies with improved efficacy against infectious diseases and cancer.

What are the emerging research areas regarding population-specific diversity of IGHG3 and its functional implications?

Several emerging research areas focus on population-specific diversity of IGHG3 and its functional implications:

  • Population genetics and evolution: Studies have revealed striking differences in IGHG3 polymorphism between population groups. Individuals of African descent show higher IGHG3 diversity compared to individuals of European descent. These differences may reflect evolutionary adaptations to distinct pathogen exposures .

  • Disease susceptibility correlations: Ongoing research is exploring how IGHG3 hinge length variations may correlate with susceptibility to various diseases. For example, one study found that IGHG3 hinge length variation was associated with the risk of critical COVID-19 .

  • Vaccine response prediction: IGHG3 polymorphisms may influence vaccine responses across populations. IgG3 responses have been shown to correlate with partial protection in HIV vaccine trials, suggesting that IGHG3 genotyping could help predict vaccine efficacy in different populations .

  • Precision medicine applications: Understanding individual IGHG3 genotypes could guide personalized therapeutic approaches:

    • Selection of appropriate antibody therapeutics based on patient IGHG3 profile

    • Prediction of response to antibody-based treatments

    • Development of population-specific antibody formulations

  • Comprehensive allotype characterization: Advanced sequencing technologies are enabling more detailed characterization of IGHG3 allotypes, revealing greater levels of allelic polymorphism than previously described through serological methods. This molecular definition provides insights into functional variations that were previously indistinguishable .

PopulationIGHG3 CharacteristicPotential Functional Implication
South AfricansExtremely low amino acid variation in IGHG1May impact antibody responses to endemic pathogens
African AmericansHigher IGHG2 and IGHG3 diversityPotentially broader range of pathogen recognition
European AmericansIntermediate IGHG3 diversityDifferent effector function profiles compared to African populations

This emerging field represents an important frontier in understanding how genetic diversity impacts immune responses across human populations.

What are the recommended protocols for characterizing IGHG3 allotypes in research populations?

For comprehensive characterization of IGHG3 allotypes in research populations, the following protocols are recommended:

  • DNA extraction and amplification:

    • Extract genomic DNA from peripheral blood or saliva samples

    • Design primers specific for IGHG3 to avoid amplification of other highly homologous IGHG genes

    • Amplify IGHG3 genomic fragments including exons encoding the CH1, hinge, CH2, and CH3 domains

  • Hinge exon analysis:

    • For hinge region analysis, use primers outside the ~200 bp repetitive element

    • Label forward primers with fluorochrome (e.g., 5-FAM) to facilitate detection

    • Determine the number of exons based on electrophoretically-defined PCR fragment length using capillary electrophoresis

  • Sanger sequencing:

    • Sequence the PCR products using Sanger methodology to identify polymorphic sites

    • Note that SNPs in IGHG3 hinge exons may be difficult to analyze directly when there is heterozygosity for exon copy number

  • Next-generation sequencing approaches:

    • For large population studies, consider targeted next-generation sequencing

    • Compare data with reference databases such as the 1000 Genomes Project, but be aware of potential inaccuracies in public databases

  • Allotype assignment:

    • Determine allotypes based on amino acid sequence variations at key positions

    • Consider both serologically defined allotypes (G3m markers) and molecular polymorphisms

    • Analyze linkage disequilibrium patterns between SNPs to define haplotypes

  • Population genetics analysis:

    • Calculate allele frequencies and test for Hardy-Weinberg equilibrium

    • Perform population differentiation tests (e.g., FST statistics)

    • Analyze patterns of linkage disequilibrium to identify selection signatures

This comprehensive approach enables accurate characterization of IGHG3 diversity across populations and forms the foundation for functional studies of allotype-specific effects.

What experimental systems are optimal for studying IgG3-mediated intracellular immunity?

To effectively study IgG3-mediated intracellular immunity, several experimental systems have proven valuable:

  • Adenovirus infection models:

    • The Ad5 system is particularly useful as it allows for clear measurement of antibody-dependent neutralization

    • Ad5-mCherry provides a fluorescent readout that can be quantified by flow cytometry

    • Wild-type and TRIM21 knockout cell comparisons allow for specific attribution of neutralization to TRIM21-mediated mechanisms

  • Cell line selection:

    • 293T cells lacking surface FcγRs are useful for isolating intracellular immunity effects

    • TRIM21 knockout cells generated using CRISPR-Cas9 serve as essential controls

    • Reporter cell lines (e.g., NF-κB reporter cells) enable measurement of immune signaling responses

  • Antibody engineering platforms:

    • Systems for generating matched antibody subclasses with identical variable regions

    • Hinge engineering capabilities for creating variants with different lengths and flexibilities

    • Site-directed mutagenesis to introduce specific amino acid changes (e.g., H433A substitution that abrogates TRIM21 binding)

  • Readout technologies:

    • Flow cytometry for quantifying viral infection levels

    • Luciferase-based reporter assays for NF-κB activation measurement

    • Western blotting for assessing complement component activation (e.g., C4 cleavage)

  • Molecular interaction studies:

    • Surface plasmon resonance for measuring binding kinetics between TRIM21 and antibody Fc regions

    • ELISA for assessing antibody binding to viral particles

    • Analytical ultracentrifugation for determining molecular interactions in solution

Using these experimental systems in combination provides comprehensive insights into the mechanisms and functional significance of IgG3-mediated intracellular immunity, particularly its enhanced capacity compared to other IgG subclasses.

How do the structural and functional properties of IgG3 compare to other IgG subclasses?

The following table summarizes key structural and functional differences between IgG3 and other IgG subclasses:

PropertyIgG1IgG2IgG3IgG4
Serum concentrationHighest (~9 mg/ml)Second (~3 mg/ml)Third (~1 mg/ml)Lowest (~0.5 mg/ml)
Hinge lengthMedium (~15 aa)Short (~12 aa)Long (32-62 aa)Medium (~15 aa)
Hinge flexibilityModerateLowHighModerate
Half-life21 days21 days7 days (R435) or 21 days (H435)21 days
TRIM21 bindingGoodGoodGoodGood
TRIM21-mediated neutralizationModerateLowHighVery low
NF-κB activation via TRIM21ModerateLowHighVery low
C1q bindingStrongWeakStrongestVery weak/none
Complement activationGoodPoorExcellentPoor
FcγR bindingGoodPoorStrongPoor

Key functional differences:

  • Intracellular immunity: IgG3 demonstrates superior capacity to mediate TRIM21-dependent virus neutralization and immune signaling compared to other subclasses, despite equal binding to TRIM21 in biochemical assays .

  • Complement activation: IgG3 is the most potent subclass for complement activation, followed by IgG1 and IgG2, while IgG4 shows little to no activity. This hierarchy mirrors C1q binding capabilities .

  • Temporal expression patterns: IgG3 responses typically occur acutely during infection, acting before IgG1 responses become dominant. This suggests a specialized role for IgG3 during initial stages of pathogen encounter .

  • Physical properties: IgG3 demonstrates poorer conformational and colloidal stability compared to IgG1, with elevated solution viscosity at high concentrations .

These distinctive properties of IgG3 likely evolved to provide specialized immune functions that complement the roles of other IgG subclasses in the adaptive immune response.

What methodological approaches can address conflicting data regarding IgG3 effector functions?

Researchers may encounter conflicting data regarding IgG3 effector functions due to various factors including allotypic variations, experimental conditions, and measurement techniques. The following methodological approaches can help address these conflicts:

  • Standardization of antibody reagents:

    • Use genetically defined IgG3 variants with known allotypes

    • Create matched IgG subclass panels with identical variable regions

    • Characterize post-translational modifications, especially glycosylation patterns

    • Verify antibody integrity and homogeneity before functional tests

  • Comprehensive allotype characterization:

    • Sequence antibody Fc regions to identify exact allotypic variants

    • Consider hinge length variations when interpreting functional data

    • Account for key amino acid positions known to affect function (positions 291, 292, 296, 435)

  • Multi-parameter functional assays:

    • Employ multiple complementary assays for each effector function

    • Include appropriate positive and negative controls

    • Use dose-response curves rather than single-concentration measurements

    • Consider kinetic measurements rather than endpoint readings

  • Systematic comparison approach:

    • Use domain swap experiments (e.g., hinge region exchanges between IgG subclasses)

    • Create point mutations to isolate the effect of specific amino acid differences

    • Develop structure-function correlations through systematic variant testing

  • Context-dependent evaluation:

    • Test effector functions in relevant cellular contexts

    • Consider target antigen density and distribution

    • Evaluate the impact of environmental factors (pH, ionic strength)

    • Assess functions across different effector cell types

  • Statistical robustness:

    • Perform multiple independent experiments

    • Use sufficient technical and biological replicates

    • Apply appropriate statistical tests for data analysis

    • Consider meta-analysis approaches when comparing across studies

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 2025 TheBiotek. All Rights Reserved.