LRRC32 antibodies are designed to target the extracellular or intracellular domains of the LRRC32 protein, a type I transmembrane glycoprotein. Key characteristics include:
Structure: Comprises 22 leucine-rich repeats (LRRs), a transmembrane domain, and a short cytoplasmic tail .
Function: Binds latency-associated peptide (LAP) of TGF-β, tethering latent TGF-β to Treg surfaces for controlled activation .
LRRC32 antibodies are widely used in:
Western Blot: Detect LRRC32 in lysates (e.g., mouse spleen cells ).
Immunohistochemistry: Localize LRRC32 in tissues (e.g., human tonsillitis ).
Functional Studies: Investigate TGF-β activation and Treg suppression mechanisms .
Treg Stability:
Transcriptional Regulation:
Therapeutic Implications:
Cross-Reactivity: Some antibodies (e.g., AF6229) show partial cross-reactivity with human LRRC32 .
Dilution Optimization: Protocols vary; e.g., 1:200–1:1000 for WB vs. 0.1 µg/mL for mouse spleen lysates .
Controls: Isotype-matched antibodies (e.g., Rat IgG1 ) are critical for flow cytometry validation.
LRRC32/GARP is a transmembrane glycoprotein of 662 amino acids with its extracellular portion containing 20 leucine-rich repeats . It functions as a cell surface receptor primarily on regulatory T-lymphocytes (Treg cells), platelets, hepatic stellate cells, and certain cancer cells . LRRC32 has been demonstrated as a Treg-specific activation marker and plays a critical role in the surface expression of latent TGF-β by binding to the complex . This protein is particularly significant in immunological research because it helps identify potent regulatory T cell subsets - LRRC32+ Tregs demonstrate greater suppressive capacity than LRRC32- Tregs . Understanding LRRC32 expression and processing provides insight into Treg function and potential immunotherapeutic strategies targeting these cells . The protein's selective expression on activated FOXP3+ regulatory T cells makes it a valuable marker for distinguishing functionally suppressive Tregs from other T cell populations .
Sample preparation depends on whether you are detecting surface or intracellular LRRC32:
For surface expression detection in flow cytometry:
Harvest cells and wash in PBS containing 1% BSA
Block non-specific binding with appropriate isotype controls
Stain with anti-LRRC32 antibody without permeabilization
For intracellular detection:
First stain surface antigens
Fix and permeabilize cells using a fixation permeabilization kit
Block with isotype control antibody (e.g., 2.5 μg/ml of IgG2b) for 30 minutes to reduce non-specific binding
Incubate with labeled anti-LRRC32 antibody
For Western blot detection:
Lyse cells in appropriate buffer (e.g., RIPA buffer)
Reduce samples before loading (LRRC32 appears at approximately 80-85 kDa on gels)
Use Immunoblot Buffer Group 1 for optimal results
When working with tissue sections for IHC, heat-induced epitope retrieval is crucial for optimal antibody binding, as demonstrated in mouse spleen tissue studies .
Differentiating between intracellular and surface expression of LRRC32 requires specific experimental approaches:
For flow cytometry:
Surface detection: Stain unfixed, non-permeabilized cells with anti-LRRC32 antibody
Intracellular detection: After surface marker staining, fix and permeabilize cells before LRRC32 staining
Compare the same cell population with both staining approaches to determine localization patterns
Studies have demonstrated that low levels of LRRC32 are present intracellularly in freshly isolated Tregs prior to activation, while surface expression occurs following activation . To accurately distinguish these populations, include appropriate controls and carefully analyze co-expression with other markers like CD25 and FoxP3 .
Microscopy techniques can also help determine localization:
Immunofluorescence microscopy shows cytoplasmic localization in certain cell types like the bEnd.3 mouse endothelioma cell line
Surface expression can be confirmed using non-permeabilized cells with confocal microscopy
Comparing signal intensity between permeabilized and non-permeabilized samples helps quantify the relative distribution
Importantly, signal peptide cleavage is essential for surface expression of LRRC32, so mutations or inhibition of this process would prevent surface localization while potentially preserving intracellular expression .
For optimal Western blot detection of LRRC32, follow these guidelines:
When detecting LRRC32 in transfected samples, note that the protein may appear at a different molecular weight (~100 kDa) in Fc fusion constructs compared to the native protein (~72 kDa) . This size discrepancy should be considered when interpreting results from recombinant proteins versus endogenous expression .
For optimal sensitivity, protein concentration should be determined and standardized across lanes, and exposure times should be optimized to avoid signal saturation while maintaining clear band visualization .
Selecting the appropriate LRRC32 antibody depends on your specific research application, target species, and experimental goals:
Consider target species reactivity:
Match antibody type to application:
Evaluate epitope binding region:
Review validated applications in literature:
Some antibodies have been cited in published research for specific applications
Consider antibodies with demonstrated performance in your experimental system
Check if the antibody detects both glycosylated and non-glycosylated forms if relevant
Match host species to your experimental system:
Always perform proper validation experiments including positive and negative controls to ensure the antibody performs as expected in your specific experimental conditions.
LRRC32 (GARP) plays a critical role in tethering latent TGF-β on the surface of activated regulatory T cells, making it an excellent target for investigating TGF-β pathways . Here are methodological approaches using LRRC32 antibodies:
Co-localization studies:
Use dual immunofluorescence with anti-LRRC32 and anti-LAP (latency-associated peptide) antibodies
Analyze co-localization patterns using confocal microscopy
Quantify Pearson's correlation coefficients between signals
Functional blockade experiments:
Use anti-LRRC32 antibodies to block the interaction between LRRC32 and latent TGF-β
Compare TGF-β signaling activities (e.g., Smad phosphorylation) in blocked versus unblocked conditions
Assess downstream functional effects on target cell populations
Pull-down and co-immunoprecipitation:
Flow cytometry analysis:
Measure surface expression of LRRC32 and LAP-TGF-β using specific antibodies
Correlate expression levels with Treg suppressive function
Sort LRRC32+ and LRRC32- populations to compare TGF-β production and activity
This approach provides mechanistic insights into how LRRC32 contributes to Treg function through TGF-β presentation, as research has demonstrated that LRRC32 binds directly to TGF-β LAP and tethers latent TGF-β on activated Treg cell surfaces, which contributes to their suppressive capacity .
To comprehensively study LRRC32's role in regulatory T cell function, researchers can employ these methodological strategies:
Comparative functional analysis of LRRC32+ and LRRC32- Treg populations:
Phenotypic characterization:
Activation-dependent expression studies:
Monitor LRRC32 expression before and after Treg activation
Correlate expression levels with suppressive function
Investigate intracellular versus surface localization during activation stages
Signal peptide processing analysis:
Knockdown/inhibition experiments:
These approaches should be combined with appropriate controls, including isotype antibody controls for flow cytometry and comparison with effector T cell populations that do not express LRRC32 .
When encountering inconsistent or weak LRRC32 detection, consider these methodological troubleshooting approaches:
For Western blot inconsistencies:
Verify protein loading: LRRC32 has a calculated molecular weight of 72 kDa but is observed at ~80-85 kDa due to glycosylation
Ensure reducing conditions: Use proper reducing agents in sample buffer
Optimize antibody concentration: Test dilution ranges from 1:200-1:1000
Check positive controls: Use HUVEC cells or mouse spleen non-B cells as validated sources
Consider membrane type: PVDF membranes may provide better results than nitrocellulose
For immunohistochemistry weak signals:
Optimize antigen retrieval: Try both TE buffer pH 9.0 and citrate buffer pH 6.0
Increase antibody concentration: Test higher concentrations within the 1:50-1:500 range
Extend incubation time: Consider overnight incubation at 4°C
Use amplification systems: Consider tyramide signal amplification for weak signals
Verify positive tissue controls: Human tonsillitis tissue and mouse spleen have confirmed expression
For flow cytometry challenges:
Block non-specific binding: Pre-incubate with isotype control (2.5 μg/ml IgG2b)
Consider activation status: LRRC32 expression increases after Treg activation
Check for intracellular expression: Some cells may have primarily intracellular rather than surface expression
Optimize permeabilization: Different fixation/permeabilization protocols may affect epitope accessibility
Use fresh samples: LRRC32 expression may decrease with extended cell culture
General considerations:
Antibody storage: Ensure proper storage at -20°C for long-term stability
Sample handling: Minimize freeze-thaw cycles for both antibodies and protein samples
Consider epitope accessibility: Previous studies using antibodies against amino acids 296-308 failed to detect LRRC32 in certain contexts
Test multiple antibody clones: Different antibodies may recognize different epitopes with varying accessibility
Always titrate antibodies in each testing system for optimal results, as recommended by manufacturers .
While LRRC32 is primarily studied in Tregs, investigating its role in non-immune cells requires specific methodological considerations:
For endothelial cells and megakaryocytes:
Verify baseline expression using Western blot on HUVEC cells, a validated positive control
Use immunofluorescence for localization studies, as demonstrated in bEnd.3 mouse endothelioma cells
Apply 10 μg/mL antibody concentration for 3 hours at room temperature for optimal staining
Counterstain with DAPI to visualize nuclei and confirm cytoplasmic localization
For platelet studies:
Use flow cytometry with careful gating strategies for these small cellular fragments
Compare expression patterns with activation markers to determine correlation with platelet activation status
Implement appropriate platelet isolation protocols to minimize activation during preparation
For cancer cells:
Screen cell lines for expression using Western blot before detailed studies
Compare expression levels with paired normal tissues when available
Correlate with TGF-β signaling activity using reporter assays
Consider analyzing LRRC32 expression in relation to tumor immunosuppressive mechanisms
For hepatic stellate cells:
Use co-staining with stellate cell markers for verification
Analyze expression changes during stellate cell activation and fibrosis progression
Consider both surface and intracellular localization patterns
Correlate with TGF-β production and fibrogenic activity
In all non-immune cell studies, appropriate controls are essential:
Include positive controls (Tregs or HUVEC cells) alongside test samples
Use multiple detection methods to confirm expression (Western blot, flow cytometry, immunostaining)
Verify specificity using competing peptides or knockout/knockdown approaches when possible
Consider context-specific expression regulation that may differ from immune cells
Optimal antibody dilution varies by application, sample type, and specific antibody clone. Here's a methodological approach to optimization:
For systematic optimization:
Western blot:
Immunohistochemistry/Immunofluorescence:
Flow cytometry:
Remember that optimal dilutions may need readjustment when:
Switching between fresh and fixed tissues
Moving between different detection systems
Working with samples from different species
Using new antibody lots
As noted in the product information, "It is recommended that this reagent should be titrated in each testing system to obtain optimal results" .
Several critical factors significantly impact LRRC32 detection in tissue sections:
Antigen retrieval method:
Fixation parameters:
Antibody selection and concentration:
Detection system selection:
Tissue-specific considerations:
Signal specificity verification:
Include negative controls (isotype antibody or secondary-only staining)
Use tissues known to lack LRRC32 expression as negative controls
Consider blocking peptide controls to confirm specificity
These factors should be systematically optimized when establishing LRRC32 detection protocols for tissue sections, with special attention to antigen retrieval methods which appear particularly critical for successful staining .
LRRC32 antibodies offer powerful tools for identifying and isolating therapeutically relevant Treg populations through these methodological approaches:
Flow cytometry-based identification and sorting:
Functional verification of isolated populations:
Assess suppressive capacity in mixed lymphocyte reaction assays
Measure TGF-β production and presentation capacity
Evaluate stability of FoxP3 expression and suppressive phenotype
Compare gene expression profiles between LRRC32+ and LRRC32- Treg subsets
Therapeutic applications:
Enrich for LRRC32+ Tregs in adoptive transfer protocols to enhance efficacy
Use LRRC32 as a quality control marker for manufactured Treg products
Monitor LRRC32 expression during ex vivo expansion to track functional potential
Correlate clinical outcomes with LRRC32 expression levels in administered Tregs
Advanced isolation strategies:
Combine magnetic pre-enrichment of CD4+CD25+ cells with LRRC32-based FACS sorting
Develop GMP-compliant isolation protocols using clinical-grade antibodies
Implement closed-system isolation platforms for therapeutic applications
Consider activation-induced expression for expanding LRRC32+ populations
This approach helps "select for more potent Treg populations" as suggested by research demonstrating that "LRRC32 surface expression may be useful as a marker that selects for more potent Treg populations" .
LRRC32/GARP antibodies present unique opportunities and challenges for cancer immunotherapy development:
Target validation strategies:
Assess LRRC32 expression in tumor-infiltrating Tregs versus peripheral Tregs
Analyze correlation between LRRC32+ Treg infiltration and patient outcomes
Determine LRRC32 expression on tumor cells themselves, as some cancer cells express LRRC32
Evaluate relationship between LRRC32 expression and TGF-β signaling in the tumor microenvironment
Therapeutic antibody development considerations:
Select antibodies binding specific epitopes that block TGF-β presentation
Evaluate different antibody formats (IgG1, IgG4, F(ab')2) for optimal efficacy
Consider Fc engineering to enhance or suppress effector functions depending on mechanism of action
Test for cross-reactivity between human and mouse LRRC32 (~80% amino acid sequence identity) for translational studies
Combination therapy approaches:
Test LRRC32-targeting agents with immune checkpoint inhibitors
Combine with TGF-β pathway inhibitors for potential synergistic effects
Evaluate sequential versus concurrent administration strategies
Monitor changes in tumor-infiltrating lymphocyte populations following treatment
Monitoring and biomarker development:
Use flow cytometry to track LRRC32+ Tregs during treatment
Develop immunohistochemistry protocols for FFPE tumor samples
Correlate LRRC32 expression with other immunosuppressive markers
Monitor TGF-β activity as a pharmacodynamic endpoint
Preclinical model selection:
These methodological considerations address the complex role of LRRC32 in regulating TGF-β bioavailability in the tumor microenvironment, which may contribute to immunosuppression and tumor progression through Treg-mediated suppression mechanisms .