B3GNT3 antibodies target the B3GNT3 protein (UDP-GlcNAc:βGal β-1,3-N-acetylglucosaminyltransferase 3), a Golgi-localized enzyme involved in synthesizing poly-N-acetyllactosamine chains and modulating glycosylation of cell surface proteins like PD-L1 . These antibodies are critical for detecting B3GNT3 in experimental workflows such as:
B3GNT3 antibodies have been extensively used to study its oncogenic roles:
B3GNT3 antibodies help explore its immunosuppressive effects:
In pancreatic cancer, B3GNT3 overexpression inversely correlates with macrophage infiltration (CD68+ cells) .
B3GNT3 modulates PD-1/PD-L1 interactions in triple-negative breast cancer, affecting T-cell activity .
In pancreatic adenocarcinoma (PAAD), high B3GNT3 expression predicts poor survival (HR = 1.89, P < 0.001) .
B3GNT3 promotes metastasis in ESCA by regulating RhoA/RAC1 signaling .
Knockdown Effects: Silencing B3GNT3 reduces proliferation, invasion, and epithelial-mesenchymal transition (EMT) in pancreatic cancer cells .
Immune Modulation: B3GNT3 expression negatively correlates with CD8+ T-cell infiltration in LUAD and PAAD .
B3GNT3 (beta-1,3-N-acetylglucosaminyltransferase 3) is a glycosyltransferase enzyme involved in the synthesis of poly-N-acetyllactosamine. It demonstrates activity for type 2 oligosaccharides and functions as a core1-1,3-N-acetylglucosaminyltransferase (Core1-beta3GlcNAcT) to form the 6-sulfo sialyl Lewis x on extended core1 O-glycans . This enzyme plays critical roles in multiple biological processes including cell adhesion, immune regulation, and cancer progression. The protein has a predicted molecular weight of approximately 43 kDa .
Detection of B3GNT3 in tissue samples typically employs these methodologies:
Immunohistochemistry (IHC): Used to localize B3GNT3 in tissue sections, with immunoreactivity primarily detected in the cytoplasm. Published studies have employed various scoring systems based on staining intensity (weak, moderate, strong) .
ELISA: Particularly useful for serum samples, as demonstrated in studies measuring B3GNT3 levels in patients with lung adenocarcinoma versus healthy controls .
Western Blotting: Commercial antibodies for B3GNT3 (such as ab96267 and ab70156) are validated for western blot applications, typically using 10% SDS-PAGE with dilutions around 1/1000 .
qRT-PCR: For quantifying B3GNT3 mRNA expression levels, as implemented in studies examining B3GNT3 expression in cancer cell lines .
To ensure antibody specificity:
Positive and negative controls: Include cell lines with known B3GNT3 expression levels. The 293T cell line has been validated for western blot applications with anti-B3GNT3 antibody .
Knockdown validation: Implement siRNA or shRNA to create B3GNT3 knockdown controls and verify reduced antibody signal. This approach has been demonstrated in studies examining B3GNT3 function in esophageal and lung cancer cell lines .
Western blot band verification: Confirm that detected bands match the predicted molecular weight of 43 kDa for B3GNT3 .
Cross-reactivity assessment: Test antibody reactivity across multiple relevant human tissues and cell lines to establish specificity patterns.
B3GNT3 expression demonstrates consistent associations with clinical outcomes across multiple cancer types:
Notably, high B3GNT3 expression serves as an independent prognostic factor in multiple cancer types, with Kaplan-Meier analyses consistently showing shorter survival times in patients with elevated expression .
Several complementary methodologies can be employed:
Bioinformatic analysis:
TIMER (Tumor Immune Estimation Resource) tool can analyze correlations between B3GNT3 expression and immune infiltration levels, particularly CD4+ T cells, neutrophils, macrophages, and dendritic cells .
TISIDB (Tumor-Immune System Interaction Database) can examine correlations between B3GNT3 and various immune signatures, including immune checkpoint gene sets .
Co-expression studies:
Functional validation:
B3GNT3 knockdown experiments followed by immune cell co-culture assays.
Analysis of cytokine production and immune checkpoint expression after B3GNT3 manipulation.
Research has identified negative correlations between B3GNT3 expression and immune infiltration levels in pancreatic cancer, particularly with macrophages (correlation coefficient = -0.366) . This suggests immunomodulatory functions that warrant further investigation.
To rigorously investigate the miR-149-5p/B3GNT3 regulatory axis:
Bioinformatic prediction and validation:
Expression analysis:
Quantify miR-149-5p and B3GNT3 expression levels in matched cancer tissues and cell lines using qRT-PCR.
Assess correlation patterns between miR-149-5p and B3GNT3 across patient samples.
Functional studies:
Transfect cancer cells with miR-149-5p mimics or inhibitors and measure changes in B3GNT3 expression using Western blot and qRT-PCR.
Perform rescue experiments where B3GNT3 is overexpressed in miR-149-5p-overexpressing cells to determine if B3GNT3 restoration reverses miR-149-5p-mediated effects.
Phenotypic assays:
Previous research has validated that miR-149-5p negatively regulates B3GNT3 expression by directly targeting its 3'-UTR, and overexpression of miR-149-5p can antagonize the tumorigenic effects of B3GNT3 in lung cancer cells .
To comprehensively investigate B3GNT3's role in invasion and metastasis:
In vitro invasion assays:
Migration analysis:
Conduct wound healing/scratch assays to measure collective cell migration.
Employ live-cell imaging to track individual cell movement patterns and dynamics.
Molecular mechanism investigation:
Analyze expression of epithelial-mesenchymal transition (EMT) markers following B3GNT3 manipulation.
Investigate changes in cell adhesion molecules and matrix metalloproteinases.
Examine the potential interaction between B3GNT3 and PD-L1, as this has been implicated in triple-negative breast cancer .
In vivo metastasis models:
Utilize tail vein injection or orthotopic implantation models with B3GNT3-modified cells to assess metastatic potential.
Employ bioluminescence imaging to track metastatic spread longitudinally.
Research has demonstrated that knockdown of B3GNT3 significantly suppresses lung cancer cell growth and invasion in vitro, and inhibits tumor development in xenograft models . Additionally, in esophageal cancer cells, B3GNT3 silencing reduced growth rate and Ki-67 expression levels .
Several technical challenges require specific methodological approaches:
Glycan structure analysis:
Mass spectrometry techniques (MALDI-TOF, LC-MS/MS) for detailed glycan profiling.
Lectin microarrays to identify differential glycosylation patterns in B3GNT3-modified cells.
Specialized glycan labeling strategies to track altered O-glycan structures.
Functional redundancy assessment:
Multi-gene knockdown approaches to address potential compensatory mechanisms from other glycosyltransferases.
CRISPR-Cas9 gene editing to create complete B3GNT3 knockout models.
Specific glycosylation target identification:
Microenvironmental context:
3D organoid cultures to better recapitulate physiological glycosylation patterns.
Co-culture systems to assess how B3GNT3-mediated glycosylation affects tumor-stromal interactions.
Current research indicates that B3GNT3 plays dual roles in cancer, with both tumor-promoting effects in various epithelial cancers and potentially tumor-suppressive effects in neuroblastoma , highlighting the complexity of glycosylation biology in different tumor contexts.
When designing experiments across different tissue types:
Baseline expression profiling:
Tissue-specific optimization:
Adjust antibody concentrations and incubation times based on known expression levels (e.g., higher dilutions for tissues with elevated B3GNT3 expression).
Optimize antigen retrieval methods specifically for each tissue type being analyzed.
Context-specific interpretation:
Consider the biological context of each tissue when interpreting B3GNT3 staining patterns.
In lung adenocarcinoma, B3GNT3 expression correlates with clinical stage progression .
In pancreatic cancer, B3GNT3 expression shows negative correlation with immune cell infiltration .
In esophageal cancer, high B3GNT3 expression is associated with poorer tissue differentiation .
Specialized protocols:
For rigorous comparative analysis:
Sample collection and processing:
Collect matched tumor and adjacent normal tissues (at least 2 cm from tumor margin).
Use consistent fixation protocols (e.g., 10% neutral buffered formalin for 24 hours) to ensure comparable antigen preservation.
IHC methodology:
Quantification approaches:
Digital image analysis with tissue segmentation for objective quantification.
Blinded assessment by multiple pathologists to reduce scoring bias.
Controls and normalization:
Include internal positive controls (tissues known to express B3GNT3).
Normalize expression against housekeeping genes when using qRT-PCR.
Multi-modal validation:
Research has demonstrated that B3GNT3 expression is significantly higher in tumor tissues compared to adjacent normal tissues across multiple cancer types, with particularly well-documented differences in lung adenocarcinoma and esophageal cancer .
To assess B3GNT3's potential as a therapeutic target:
Target validation strategies:
Implement inducible shRNA or CRISPR-Cas9 systems to modulate B3GNT3 expression in established tumors.
Assess changes in tumor growth, metastasis, and immune infiltration following B3GNT3 inhibition.
Small molecule screening:
Develop high-throughput assays to identify inhibitors of B3GNT3 enzymatic activity.
Test candidate compounds in cell-based glycosylation assays to confirm target engagement.
Combination therapy assessment:
Biomarker development:
Establish standardized protocols for measuring serum B3GNT3 levels as a clinical biomarker.
Develop companion diagnostics to identify patients most likely to benefit from B3GNT3-targeted therapies.
Research has demonstrated that inhibiting B3GNT3 while simultaneously inhibiting PD-L1 significantly increases apoptosis and inhibits proliferation in lung adenocarcinoma models . Additionally, overexpression of miR-149-5p, which negatively regulates B3GNT3, antagonizes tumorigenic effects in vitro , suggesting multiple therapeutic intervention points.
For comprehensive analysis of B3GNT3-immune checkpoint interactions:
Co-expression and co-localization studies:
Perform multiplex immunofluorescence for B3GNT3 and immune checkpoint proteins (PD-1, PD-L1, CTLA-4).
Analyze spatial relationships and potential co-regulation in tissue microarrays.
Glycosylation analysis of checkpoint proteins:
Employ immunoprecipitation followed by glycoproteomic analysis to identify B3GNT3-dependent glycan modifications on immune checkpoint proteins.
Utilize site-directed mutagenesis to eliminate specific glycosylation sites and assess functional consequences.
Functional interaction assays:
Implement B3GNT3 knockdown/overexpression followed by immune checkpoint blockade to assess combinatorial effects.
Conduct T-cell killing assays to determine how B3GNT3 modulation affects tumor cell susceptibility to immune attack.
Mechanistic pathway analysis:
Investigate downstream signaling events following B3GNT3-mediated modifications of immune checkpoint proteins.
Employ phospho-proteomics to map altered signaling networks.
Research has established that B3GNT3 is essential for epidermal growth factor-induced communication between PD-1 and PD-L1 in triple-negative breast cancer . The TISIDB analysis has also revealed correlations between B3GNT3 expression and immune checkpoint markers such as ICOS and NECTIN2 (CD112) , suggesting broad implications for immunotherapy response.