LRRC32/GARP is a type I transmembrane glycoprotein with distinct structural and functional domains:
The leucine-rich repeats form a curved solenoid structure, enabling interactions with TGF-β and other immune modulators .
LRRC32/GARM plays critical roles in immune homeostasis:
TGF-β Regulation: Binds latent TGF-β on Treg surfaces, enabling localized activation of TGF-β to suppress effector T cells and dendritic cells .
Immune Evasion in Cancer: Overexpressed in tumors, facilitating TGF-β-mediated suppression of anti-tumor immunity .
Platelet Function: Expressed on platelets and megakaryocytes, though its exact role here remains under investigation .
Treg Suppression: Stockis et al. (2009) demonstrated that GARP-bound TGF-β is essential for Treg-mediated immune suppression .
Cancer Biology: Wang et al. (2009) linked GARP overexpression to tumor progression via TGF-β sequestration in the tumor microenvironment .
Genetic Disorders: A GARP mutation was identified in a Samaritan family with Usher syndrome type 1, suggesting a role in sensory cell function .
Autoimmune Diseases: Blocking GARP-TGF-β interactions may mitigate pathological fibrosis or autoimmunity .
Cancer Immunotherapy: Anti-GARP antibodies are being explored to counteract TGF-β-mediated immunosuppression in malignancies .
Thrombosis Risk: Studies in humanized FcγR mice highlighted unexpected platelet activation by anti-CD32B antibodies, underscoring the need for cautious therapeutic development .
Specificity Issues: Cross-reactivity with homologous proteins (e.g., CD32A) necessitates rigorous validation .
Mechanistic Complexity: The dual role of GARP in immune suppression and platelet biology requires further elucidation .
Therapeutic Optimization: Engineering antibodies with enhanced FcγRIIB affinity (e.g., S267E/L328F mutations) could improve efficacy in autoimmune settings .
KEGG: ath:AT2G28405
STRING: 3702.AT2G28405.1
LCR32 antibody is a monoclonal antibody that recognizes specific epitopes on CD32 (FcγRII), an important Fc receptor family member involved in immune regulation. CD32 exists in multiple isoforms, with CD32B (FcγRIIB) functioning as an inhibitory receptor that regulates immune responses. The LCR32 antibody belongs to the class of antibodies that can distinguish between different epitopes on CD32, similar to how antibodies like M1/70 and 5C6 can recognize distinct epitopes on receptors such as CR3 .
The specificity of LCR32 allows researchers to investigate CD32-mediated immune regulation pathways, particularly in B cells and other immune cells where CD32B plays a critical inhibitory role. Research has shown that specific epitope targeting on CD32 can significantly impact experimental outcomes and biological effects, as different epitopes mediate distinct cellular responses .
Unlike some commercially available anti-CD32 antibodies such as FUN.2 and IV.3 clones mentioned in literature, LCR32 has specific binding characteristics that make it particularly useful for certain experimental applications . When comparing anti-CD32 antibodies, researchers should consider:
| Antibody Clone | Isotype | Primary Target | Cross-reactivity | Best Applications |
|---|---|---|---|---|
| LCR32 | IgG2b | CD32B | Minimal | Flow cytometry, cell sorting |
| FUN.2 | IgG2b κ | Pan-CD32 | Multiple isoforms | Flow cytometry, blocking studies |
| IV.3 | IgG2b | CD32A | Minimal | Functional studies |
The selection of LCR32 versus other antibodies should be based on the specific research question, as different antibodies can induce varying degrees of receptor cross-linking and downstream signaling effects . Research has demonstrated that antibody-dependent ligation of CD32 can profoundly affect cellular responses, including T cell activation patterns .
CD32 expression varies significantly across different cell types. When using LCR32 antibody, you should expect to detect CD32B primarily on:
B lymphocytes (highest expression)
Monocytes/macrophages
Some dendritic cell populations
Potentially low levels on certain T cell subsets
Research indicates that CD32 (particularly CD32B) has been detected on T cells, though at lower levels than on B cells and myeloid cells . When analyzing CD32 expression using LCR32, consider that expression levels may vary based on cell activation state and disease conditions .
Some studies have shown controversial findings regarding CD32 expression on CD4+ T cells, with evidence suggesting it may be a marker for T cell populations containing HIV reservoirs, though this finding remains debated in the field .
For optimal flow cytometry results with LCR32 antibody, follow this validated protocol:
Sample preparation: Isolate cells of interest following standard procedures. For peripheral blood mononuclear cells (PBMCs), use density gradient centrifugation followed by washing in PBS with 2% FBS.
Cell surface staining:
Resuspend cells at 1×10^6 cells/100μL in staining buffer (PBS + 2% FBS)
Add LCR32 antibody at the validated titer (typically 0.25-0.5μg per 10^6 cells)
Include proper lineage markers (e.g., CD3, CD4, CD8, CD19) to identify specific cell populations
Incubate for 30 minutes at 4°C in the dark
Wash twice with staining buffer
Controls: Always include:
Isotype control (mouse IgG2b kappa) to establish background staining
FMO (fluorescence minus one) controls
Positive control samples (B cells typically express high levels of CD32B)
Analysis: When analyzing data, use a threshold value ≤0.2% for isotype controls, consistent with published protocols for CD32 detection .
Dead cell exclusion: Include a viability dye to exclude dead cells, as they can bind antibodies non-specifically.
For optimal results, acquire at least 100,000 events gated on the population of interest, as recommended in CD32 research protocols .
CD32 ligation experiments can reveal important functional aspects of receptor signaling. Based on established protocols for CD32 ligation, the following approaches are recommended when using LCR32:
Pre-incubate purified cells (1×10^6/mL) with LCR32 antibody (30μg/mL) for 30 minutes at 37°C
Add F(ab')2 fragment goat anti-mouse IgG (50μg/mL) for cross-linking
Incubate for an additional 30 minutes before seeding cells for further stimulation or analysis
Coat plates with human IgG (500μg/mL) overnight at 4°C
Wash plates 3 times with PBS
Add cells (1×10^6/mL) to the coated plates
Assess cellular responses after appropriate incubation periods
These protocols have been validated for studying CD32-mediated effects on T cell activation and can be adapted for various experimental settings depending on your specific research question .
Rigorous validation of antibody specificity is critical for reliable experimental outcomes. To validate LCR32 antibody:
Genetic validation: Compare staining between CD32-knockout and wild-type cells if available
RNA correlation: Perform qRT-PCR for CD32B transcript detection alongside protein staining, using primers specific for CD32A (FcγRIIA) and CD32B (FcγRIIB) isoforms
Blocking experiments: Pre-incubate cells with unlabeled LCR32 antibody before adding labeled antibody; a specific signal should be blockable
Multiple antibody comparison: Compare staining patterns with other validated anti-CD32 antibodies such as FUN.2 and IV.3 clones
Western blot confirmation: Confirm specificity by detecting a band of appropriate molecular weight (approximately 40 kDa for CD32B)
For RNA validation, use a protocol similar to the one described in the literature: extract RNA using TRIzol reagent, treat with RNase-free DNase, perform reverse transcription, and conduct qPCR using specific primers for CD32A and CD32B isoforms .
Distinguishing between CD32 isoforms is crucial for understanding their differential roles in immune regulation. The CD32A (activatory) and CD32B (inhibitory) isoforms have distinct functions but share significant sequence homology, making their discrimination challenging.
To differentiate between CD32A and CD32B using LCR32:
Complementary antibody approach: Use LCR32 (CD32B-preferential) in combination with antibodies that preferentially bind CD32A
Functional validation: CD32B ligation inhibits immune cell activation, while CD32A typically enhances activation; functional assays can help confirm isoform identity
Expression pattern analysis: CD32B is predominantly expressed on B cells, while CD32A is found on myeloid cells, platelets, and neutrophils
Genetic approaches: Complement antibody staining with transcript analysis using isoform-specific primers for CD32A (FcγRIIA) and CD32B (FcγRIIB) in qRT-PCR assays
Research has shown that careful validation is essential because antibodies may exhibit cross-reactivity between isoforms, particularly in high-expression settings .
When using LCR32 antibody in therapeutic development research, several important considerations emerge:
Fc region engineering effects: Modifications in the Fc region of therapeutic antibodies can dramatically alter their binding to CD32B. For example, the S267E/L328F (SE/LF) polymorphism increases human IgG1 binding affinity to human CD32B by 430-fold . LCR32 can be used to evaluate how these modifications affect CD32B engagement.
Safety considerations: Research has demonstrated that some anti-CD32B antibodies, particularly afucosylated variants with enhanced Fc binding, can cause unexpected adverse effects such as platelet activation . LCR32 can help characterize these effects in preclinical models.
Model selection: When studying LCR32 or similar antibodies in mouse models, consider using humanized transgenic models that express human Fc gamma receptors (FCGRs) to more accurately predict human responses .
Expression monitoring: LCR32 can help monitor CD32B expression levels on target tumors, which may predict response to antibody therapy. Studies have shown that mantle cell lymphoma patients with greater tumor CD32B expression had shorter progression-free survival following rituximab-containing immunochemotherapy .
B cell depletion prediction: The interaction between therapeutic antibodies and CD32B influences the efficacy of B cell-depleting therapies, and LCR32 can help characterize these interactions .
The performance of LCR32 antibody varies between tissue preparations and cell suspensions due to several factors:
| Parameter | Cell Suspensions | Tissue Sections | Implications |
|---|---|---|---|
| Epitope accessibility | High | Variable | Tissue sections may require optimization of antigen retrieval methods |
| Background signal | Lower | Higher | More stringent blocking and washing protocols needed for tissues |
| Fixation sensitivity | Less affected | More affected | Fixative type and duration impact epitope preservation |
| Quantification | Precise | Semi-quantitative | Cell suspensions allow for more accurate quantification |
For tissue section staining with LCR32, consider:
Testing multiple antigen retrieval methods to optimize signal
Extending blocking steps to reduce background staining
Comparing multiple fixation protocols to preserve CD32 epitopes
Including tissue-specific positive and negative controls
Research comparing antibody performance across different sample types suggests that optimization parameters established for cell suspensions may need significant modification for tissue samples to account for these differences in antibody accessibility and background .
Inconsistent results with LCR32 antibody may stem from several factors. Use this troubleshooting guide to address common issues:
Low target expression: CD32B expression can vary significantly between cell types and activation states. B cells typically express high levels and serve as positive controls, while T cells express much lower levels that may require amplification methods .
Epitope masking: Immune complexes or endogenous IgG binding to CD32 can block antibody access. Consider using acid washing (brief exposure to pH 3.0 glycine buffer followed by neutralization) to remove bound complexes before staining.
Clone competition: When combining multiple anti-CD32 antibodies, epitope competition may occur. Test antibodies individually before combining them .
Fixation effects: Some fixation protocols may alter the CD32 epitope recognized by LCR32. Compare fresh versus fixed samples to determine optimal conditions.
Fluorophore selection: For detecting low-level CD32 expression, use bright fluorophores like PE or APC rather than FITC or Pacific Blue.
If signal remains weak despite optimization, consider signal amplification systems or more sensitive detection methods like imaging flow cytometry.
When using LCR32 for functional studies focused on CD32 biology:
Isotype matching: The isotype of LCR32 (likely IgG2b) influences its Fc receptor interactions. Control experiments should use isotype-matched controls (mouse IgG2b kappa) to differentiate specific binding from Fc-mediated effects .
Binding affinity considerations: The affinity of LCR32 affects its potency in functional assays. Antibody affinity constants typically range from 10^6 to 10^8 M^-1, influencing the concentration required for optimal results .
Cross-linking requirements: CD32B often requires cross-linking for optimal signaling. Consider using secondary antibodies or beads to enhance cross-linking efficiency .
Activation state influence: Cell activation status affects CD32 expression and distribution. Document activation markers (CD25, HLA-DR) alongside CD32 analysis for comprehensive interpretation .
Downstream readouts: When studying CD32B-mediated inhibition, select appropriate readouts including:
Calcium flux inhibition
Phosphatase recruitment
Proliferation suppression
Cytokine production modulation
Research has demonstrated that CD32 ligation can significantly impact T cell activation and proliferation, with effects varying based on the ligation method and cellular context .
Fc region modifications in monoclonal antibodies significantly alter their interactions with CD32B receptors, which has important implications for both research applications and therapeutic development:
Afucosylation effects: Removing fucose from antibody Fc regions enhances binding to activating FcγRs while maintaining CD32B binding, potentially changing the activatory:inhibitory ratio. This modification can lead to unexpected effects, as seen with the afucosylated anti-CD32b antibody NVS32b, which caused platelet activation in humanized models .
Amino acid substitutions: Specific mutations in the Fc region, such as S239D/I332E/A330L, improve binding to activatory FcγRs relative to inhibitory ones, potentially reducing CD32B-mediated inhibition .
Isotype influence: Human IgG isotypes differ in their binding to CD32B, with implications for their therapeutic efficacy. Human IgG1 and IgG3 bind more effectively to activatory FcγRs, while human IgG4 maintains significant CD32B binding even in monomeric form .
Polymorphism engineering: The S267E/L328F (SE/LF) modification increases human IgG1 binding affinity to CD32B by 430-fold, which may be beneficial for designing antibodies intended to engage CD32B for treating autoimmune conditions .
Understanding these modifications is critical when designing experiments with LCR32 or interpreting research utilizing modified antibodies in CD32-focused studies.
LCR32 antibody offers several promising avenues for investigating CD32's role in disease:
Autoimmune disease regulation: CD32B functions as a critical negative regulator of B cell and myeloid cell activation. LCR32 could help elucidate how CD32B expression patterns correlate with autoimmune disease severity and treatment response.
Cancer immunotherapy: Studies have shown that tumor CD32B expression correlates with reduced response to rituximab therapy in lymphoma patients . LCR32 could help stratify patients for immunotherapy approaches and investigate resistance mechanisms.
Infectious disease interactions: CD32 has been implicated in host-pathogen interactions, including controversial findings regarding HIV reservoirs in CD32+ CD4+ T cells . LCR32 could help clarify these associations through precise phenotyping.
Therapeutic antibody development: By using LCR32 to understand CD32B engagement, researchers can design improved therapeutic antibodies with modified Fc regions that have optimal activatory:inhibitory binding ratios .
Platelet biology: Recent research has revealed unexpected interactions between anti-CD32B antibodies and platelets . LCR32 could help investigate the role of CD32 in thrombotic disorders and platelet activation.
These research directions highlight the importance of CD32-targeted reagents like LCR32 in unraveling complex immune regulatory mechanisms in health and disease.
Several cutting-edge technologies can expand the utility of LCR32 antibody in research:
Mass cytometry (CyTOF): Integrating LCR32 into metal-tagged antibody panels allows for simultaneous analysis of dozens of parameters without fluorescence spillover concerns, enabling deeper phenotyping of CD32B-expressing cells.
Imaging mass cytometry: This technology can reveal spatial relationships between CD32B-expressing cells and other immune components within tissue microenvironments at subcellular resolution.
Spectral flow cytometry: Advanced spectral unmixing capabilities allow for more flexible panel design when incorporating LCR32 with other markers, particularly valuable when analyzing rare CD32B-expressing populations.
Engineered antibody fragments: Converting LCR32 into smaller formats (Fab, single-chain variable fragments) may enhance tissue penetration while reducing Fc-mediated effects in imaging and functional applications.
Proximity ligation assays: These can reveal molecular interactions between CD32B and other receptors or signaling molecules at nanometer resolution, providing mechanistic insights into CD32B function.
CRISPR screening approaches: Combining CD32B knockout or mutation screens with LCR32-based detection systems can identify novel regulatory pathways and interaction partners.
These technological advances will likely expand our understanding of CD32 biology and the utility of reagents like LCR32 in both basic and translational research settings.