LGALS3BP antibody binds to Galectin-3-binding protein (LGALS3BP), a secreted protein encoded by the LGALS3BP gene on chromosome 17q25. The protein interacts with integrins (α1β1, α5β1, αvβ1, α6β1) and galectins, influencing cellular adhesion, immune response, and tumor microenvironment (TME) dynamics . Elevated LGALS3BP levels correlate with poor prognosis in cancers and HIV infection .
LGALS3BP antibodies interfere with oncogenic signaling by blocking integrin-mediated pathways. Key mechanisms include:
| Target Pathway | Effect of Antibody Intervention | Outcome |
|---|---|---|
| Integrin α5β1 binding | Inhibits Akt/JNK/Ras-ERK signaling | Reduces survival and proliferation |
| Galectin-3 interaction | Disrupts TME crosstalk | Impedes angiogenesis and metastasis |
| Extracellular vesicle (EV) binding | Blocks EV-mediated communication | Limits tumor progression |
For example, the SP2 antibody inhibits LGALS3BP-driven integrin activation, suppressing motility and migration without compromising cell viability .
LGALS3BP antibodies are leveraged in antibody-drug conjugates (ADCs) for precision oncology:
1959-sss/DM3: Targets LGALS3BP on neuroblastoma-derived EVs, achieving an 80% reduction in lung metastases in murine models .
DM4-conjugated ADC: Demonstrates potent cytotoxicity in oral squamous cell carcinoma (OSCC) with an IC~50~ of 0.3 nM .
| Cancer Type | Model | Response |
|---|---|---|
| Neuroblastoma | Orthotopic PDX models | Tumor eradication in 60% of cases |
| OSCC | HOC621/CAL27 cell lines | 95% growth inhibition at 1 nM DM4 |
LGALS3BP antibodies aid in biomarker detection:
OSCC Diagnosis: Cytoplasmic LGALS3BP overexpression correlates with aggressive histology (p < 0.01) .
Serum Monitoring: Elevated serum LGALS3BP levels predict metastatic relapse in breast and lung cancers .
Recent studies highlight:
EV Targeting: LGALS3BP antibodies selectively bind cancer-derived EVs, enabling TME-specific drug delivery .
Immune Modulation: Antibody intervention enhances NK/LAK cell cytotoxicity by disrupting LGALS3BP-Mac-2 interactions .
LGALS3BP is a glycosylated protein with a calculated molecular weight of 65 kDa (585 amino acids), though it typically appears as 65-90 kDa in Western blots due to extensive post-translational modifications . Functionally, LGALS3BP serves as a negative regulator of NF-κB activation and proinflammatory cytokine production . Research demonstrates that it interacts with transforming growth factor β-activated kinase 1 (TAK1), inhibiting its phosphorylation and suppressing its kinase activity . In cancer biology, LGALS3BP is involved in various tumoral progression mechanisms including invasion, migration, immune evasion, and metastatic dissemination .
When designing experiments to study LGALS3BP function, researchers should consider its role both as a cellular protein and as a secreted factor that can act through autocrine and paracrine mechanisms. Immunoprecipitation approaches targeting TAK1 can help confirm the interaction with LGALS3BP in your experimental system.
LGALS3BP antibodies have been validated for multiple experimental applications including:
When selecting an antibody for your specific application, consider whether the antibody has been validated in similar sample types. The 10281-1-AP antibody, for example, has been cited in numerous publications spanning diverse applications .
LGALS3BP expression varies across tissues and is particularly elevated in cancer contexts. Expression patterns include:
Normal tissues/cells:
Human fetal brain tissue
Human milk
Human plasma
Various cell lines including HeLa, COLO 320, HEK-293, HepG2, and A549
Cancer tissues:
Glioblastoma multiforme (GBM), with significantly higher expression than normal brain tissue
Adenoid cystic carcinoma (ACC), showing heterogeneous expression patterns
When studying LGALS3BP expression, it's important to include appropriate positive controls from this list. Immunohistochemical analysis has revealed that expression can be heterogeneous within tumor samples, suggesting the need for comprehensive tissue assessment rather than limited sampling .
Optimizing antibody dilutions is critical for achieving specific signal while minimizing background. For LGALS3BP detection, recommended dilution ranges are:
Methodological considerations for optimization:
Perform a titration series around the recommended range (e.g., 1:500, 1:1000, 1:2000, 1:4000, 1:8000 for WB)
Include both positive controls (e.g., HepG2 cells) and negative controls
For IHC, test both suggested antigen retrieval methods: TE buffer pH 9.0 (preferred) or citrate buffer pH 6.0
Evaluate signal-to-noise ratio at each dilution rather than simply selecting the strongest signal
For challenging samples, consider extending incubation times rather than increasing antibody concentration to improve signal quality
Remember that optimal dilution is sample-dependent; titration in each experimental system is essential for obtaining reliable results .
When performing Western blot analysis of LGALS3BP, researchers should be aware of the following molecular weight considerations:
The discrepancy between calculated and observed weight is primarily due to extensive glycosylation
Methodological recommendations:
Use molecular weight markers that span 50-100 kDa range
Run gels with sufficient resolution in the 65-90 kDa region (8-10% polyacrylamide gels are suitable)
Be prepared for potential variation in apparent molecular weight across different sample types due to differential glycosylation patterns
For cleaner detection, consider longer wash steps after primary and secondary antibody incubations
If multiple bands appear, enzymatic deglycosylation (e.g., with PNGase F) can help confirm the specific LGALS3BP band
These considerations ensure accurate identification of LGALS3BP in Western blot applications.
LGALS3BP shows significant expression differences between normal and cancer tissues, making it a protein of interest for cancer diagnostics and therapeutics:
Cancer overexpression: LGALS3BP is "largely overexpressed in tumor tissue compared to the non-neoplastic counterpart," as demonstrated across multiple cancer types .
Specific cancer findings:
Glioblastoma multiforme (GBM): Immunohistochemical analysis shows high LGALS3BP expression compared to normal brain tissue
Adenoid cystic carcinoma (ACC): Heterogeneous expression observed in both PDX models and patient tumor samples
Multiple other cancers including melanoma and neuroblastoma show elevated expression
Methodological approaches for comparative analysis:
Paired analysis of tumor and adjacent normal tissue from the same patient
Tissue microarray (TMA) analysis across multiple patients
Quantitative image analysis of immunohistochemistry staining intensity
Correlation of protein expression with patient outcomes and clinical parameters
Vesicular vs. total protein: In GBM patients, "the amount of vesicular but not total circulating protein is increased" compared to healthy donors , suggesting the importance of analyzing both total and vesicle-associated LGALS3BP.
This differential expression pattern provides both diagnostic opportunities and therapeutic rationale for targeting LGALS3BP in cancer.
Optimal sample preparation is crucial for reliable LGALS3BP detection across different experimental applications:
For Western Blot analysis:
Lysis buffer: RIPA buffer containing protease inhibitors is suitable for most applications
For phosphorylation studies (e.g., TAK1 interaction), include phosphatase inhibitors
Process samples quickly and maintain cold temperatures to prevent protein degradation
For secreted LGALS3BP, concentrate cell culture supernatants using centrifugal filters or TCA precipitation
For Immunohistochemistry (IHC):
Fixation: 10% neutral buffered formalin fixation for 24-48 hours
Antigen retrieval: TE buffer pH 9.0 is recommended as the primary method, with citrate buffer pH 6.0 as an alternative
Blocking: Use 3-5% normal serum or BSA to reduce background
For multiplexed staining, carefully select antibodies from different host species
For Immunoprecipitation (IP):
Gentler lysis buffers (e.g., NP-40 or CHAPS-based) help preserve protein interactions
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Use 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate
For co-IP studies targeting TAK1 interactions, consider crosslinking approaches
For Extracellular Vesicle (EV) analysis:
Isolation methods: Differential ultracentrifugation, size-exclusion chromatography, or precipitation methods
Validation: Confirm EV isolation using markers like CD63, CD9, and CD81
Characterization: Nanoparticle tracking analysis and electron microscopy
Controls: Include detergent controls to confirm vesicular nature of the signal
These optimized methods will enhance detection sensitivity and specificity across applications.
Validating antibody specificity is essential for generating reliable data. For LGALS3BP antibodies, a comprehensive validation approach should include:
Genetic validation approaches:
Biochemical validation methods:
Peptide competition assays where pre-incubation with immunizing peptide abolishes specific signal
Immunoprecipitation followed by mass spectrometry to confirm identity
Deglycosylation experiments to confirm glycoprotein identity based on mobility shift
Multiple antibody approach:
Compare results using antibodies targeting different LGALS3BP epitopes
Consistent results with different antibodies increase confidence in specificity
Expression pattern validation:
Application-specific controls:
For IHC/IF: Include isotype controls and primary antibody omission controls
For WB: Include positive control lysates from cells with confirmed expression
For IP: Perform parallel IPs with non-specific IgG
Implementing these validation strategies will establish confidence in antibody specificity and experimental results.
Proper experimental controls are essential for accurate interpretation of LGALS3BP expression studies:
Positive Controls:
Cell lines: HeLa, COLO 320, HEK-293, HepG2, and A549 cells all express LGALS3BP and serve as reliable positive controls
Tissues: Human fetal brain, milk, and plasma samples have confirmed LGALS3BP expression
Cancer tissues: Esophageal, breast, and colon cancer tissues show robust expression
Negative Controls:
LGALS3BP knockout models: CRISPR-Cas9 generated LGALS3BP-knockout mice provide ideal negative controls
Antibody controls: Isotype-matched IgG or pre-immune serum in place of primary antibody
Peptide competition: Primary antibody pre-incubated with immunizing peptide
Expression Modulation Controls:
Overexpression: Transient transfection with LGALS3BP expression vectors
Knockdown: siRNA or shRNA targeting LGALS3BP with scrambled sequences as controls
LPS stimulation: For NF-κB pathway studies, compare with and without LPS treatment
Technical Controls:
Loading controls: β-actin or GAPDH for Western blots
Microscopy controls: DAPI nuclear counterstain for IHC/IF
Quantification controls: Standard curves for ELISA-based quantification
These multi-layered controls ensure reliable interpretation of LGALS3BP expression data across experimental contexts.
LGALS3BP functions as a significant negative regulator of NF-κB signaling through specific molecular mechanisms:
TAK1 interaction mechanism:
Impact on inflammatory signaling:
Experimental evidence:
Experimental approaches to study this pathway:
NF-κB reporter assays (luciferase) to measure activation
Immunoblotting for phosphorylated TAK1 and downstream components
Co-immunoprecipitation to confirm LGALS3BP-TAK1 interaction
Cytokine ELISAs and qPCR to measure inflammatory outputs
This regulatory role suggests LGALS3BP may have therapeutic potential for inflammatory conditions beyond its applications in cancer.
Developing LGALS3BP-targeted therapeutics presents several complex challenges that researchers must address:
Expression heterogeneity:
Protein characteristics challenges:
Physiological function considerations:
Technical and development challenges:
For antibody-drug conjugates, controlled conjugation chemistry and optimal drug-to-antibody ratio are critical
"Generation of stable cell line producing the engineered 1959-sss antibody as well as scale-up of the site-specific linker-less conjugation process" presents manufacturing challenges
For brain tumors, blood-brain barrier penetration must be considered
Preclinical model limitations:
Despite these challenges, promising results have been obtained with anti-LGALS3BP ADCs, showing "long-lasting response... in 100% of treated animals" in an ACC model and "potent and dose-dependent antitumor activity" in GBM models .
LGALS3BP antibodies provide valuable tools for investigating extracellular vesicle (EV) biology, particularly in cancer research:
Vesicular LGALS3BP as cancer biomarker:
"Compared with healthy donors, the amount of vesicular but not total circulating protein is increased" in glioblastoma patients
"Plasma vesicular LGALS3BP levels correlate with glioma tumour grade, making this protein a potential biomarker for early detection"
Research has shown that "LGALS3BP can be used for liquid biopsy as a marker of disease" in GBM models
Methodological approaches for EV isolation and analysis:
Differential ultracentrifugation: Sequential centrifugation steps (300g, 2000g, 10,000g, 100,000g)
Size-exclusion chromatography: Separate EVs based on size using specialized columns
Density gradient separation: Distinguish EV subpopulations with different densities
Characterization: Nanoparticle tracking analysis, electron microscopy, and Western blotting
LGALS3BP detection in EVs:
Western blotting of EV lysates using anti-LGALS3BP antibodies
ELISA-based quantification for high-throughput analysis
Immunoelectron microscopy for visualization of LGALS3BP on individual EVs
Flow cytometry using bead-based capture systems for EV analysis
Experimental design considerations:
Include EV markers (CD9, CD63, CD81) to confirm vesicle isolation
Use negative markers (GM130, calnexin) to exclude cellular contamination
Compare multiple isolation methods to ensure consistent results
Match isolation protocols between patient samples and controls
This approach allows researchers to leverage LGALS3BP as a biomarker for minimally invasive liquid biopsy applications, potentially improving early cancer detection and monitoring.
Investigating LGALS3BP's potential role in cancer immune evasion requires specialized experimental approaches:
Tumor-immune cell interaction studies:
Co-culture systems: Compare wild-type vs. LGALS3BP-knockout tumor cells with immune cells
Functional assays: Measure T cell proliferation, cytotoxicity, and cytokine production
Migration assays: Assess immune cell recruitment in response to LGALS3BP-containing conditioned media
NF-κB pathway analysis in immune contexts:
In vivo immune profiling:
Vesicular LGALS3BP immunomodulation:
Isolate LGALS3BP-containing EVs from tumor cells
Assess effects on dendritic cell maturation and T cell activation
Compare immunomodulatory properties of EVs from LGALS3BP-knockout vs. wild-type cells
Molecular and cellular analysis techniques:
These approaches can help elucidate whether LGALS3BP contributes to tumor immune evasion, potentially opening new avenues for immunotherapy combinations.
Post-translational modifications (PTMs), particularly glycosylation, significantly impact LGALS3BP antibody recognition and must be carefully considered:
Molecular weight variability due to glycosylation:
Experimental approaches to address PTM variability:
Enzymatic deglycosylation: Treat samples with PNGase F to remove N-linked glycans
Compare native vs. deglycosylated forms on Western blots
Use multiple antibodies targeting different epitopes to ensure comprehensive detection
Include positive control samples with known glycosylation patterns
Technical considerations for antibody selection:
Methodological recommendations for consistent detection:
Run gradient gels to improve resolution of different glycoforms
Consider lectin blotting in parallel to assess glycosylation patterns
For quantitative analysis, compare results from antibodies targeting different regions
Document the specific molecular weight observed in your experimental system
Therapeutic implications:
For antibody-drug conjugates like 1959-sss/DM4, PTM variability could affect targeting efficiency
Selection of antibodies for therapeutic use should prioritize epitopes consistently presented despite PTM variations
Understanding these PTM effects is crucial for reliable detection and effective therapeutic targeting of LGALS3BP.
Development of effective LGALS3BP-targeted antibody-drug conjugates (ADCs) requires sophisticated methodological approaches, as exemplified by the 1959-sss/DM4 ADC described in the literature:
Antibody engineering considerations:
Target validation methodologies:
Comprehensive immunohistochemical analysis across normal and cancer tissues
Quantitative assessment of expression levels in target cancers
Heterogeneity analysis within tumor samples to predict response rates
Confirmation that LGALS3BP is "largely overexpressed in the tumor tissue compared to the non-neoplastic counterpart"
Strategic payload selection:
Conjugation technology optimization:
Preclinical evaluation approaches:
In vivo imaging using "[89Zr]Zr-DFO-1959" radioimmunoconjugate to confirm tumor targeting
Patient-derived xenograft (PDX) models that maintain tumor heterogeneity
Efficacy studies demonstrating "durable tumor growth inhibition (TGI) in 100% of animals without observed toxicity"
Pharmacokinetic and biodistribution analysis
The 1959-sss/DM4 ADC has demonstrated impressive preclinical results across multiple cancer types including "potent and durable antitumor activity in melanoma, neuroblastoma and glioblastoma" and promising activity in adenoid cystic carcinoma , warranting further clinical development.