TBC1D3G antibodies are polyclonal reagents generated in rabbits, targeting specific epitopes of the human TBC1D3G protein. These antibodies are validated for research applications such as Western blot (WB), ELISA, and immunocytochemistry (ICC) . Key characteristics include:
Immunogen: Recombinant human TBC1D3G protein fragments (e.g., 312-549AA or 231-281AA) .
Specificity: Recognizes endogenous TBC1D3G in humans, mice, and rats .
Formulation: Liquid in PBS with 50% glycerol, 0.5% BSA, and 0.03% Proclin 300 .
TBC1D3G is a Rab5-specific GTPase-activating protein implicated in endosomal trafficking and immune modulation. Key functional and structural details include:
TBC1D3G antibodies are primarily used in:
TBC1D3G is part of the TBC1D3 family, which is linked to tumor progression and immune regulation in cancers like kidney renal clear cell carcinoma (KIRC) . Key findings include:
TBC1D3G expression correlates with immune inhibitors such as CTLA4, PD-1, and LAG3, which are therapeutic targets in cancer immunotherapy .
| Immune Marker | Correlation with TBC1D3G | p-value |
|---|---|---|
| CTLA4 | Positive | |
| PDCD1 (PD-1) | Positive | |
| CD160 | Positive |
TBC1D3G expression associates with immune cell activity in KIRC:
Research Use Only: Not validated for diagnostic or therapeutic applications .
Storage: Stable at -20°C or -80°C; avoid freeze-thaw cycles .
Further studies are needed to elucidate TBC1D3G’s role in immune evasion mechanisms and its potential as a biomarker for cancer prognosis .
TBC1D3G (TBC1 Domain Family Member 3G) functions as a GTPase-activating protein (GAP) specifically for RAB5, but does not act on RAB4 or RAB11 . It belongs to the TBC1D3 family, which is characterized by the presence of a RAB-GAP domain called the TBC1 domain . These proteins typically function as attenuators of RABs, which are considered master switches of membrane traffic. TBC1D3G is part of a hominoid-specific gene family that evolved to modulate signaling pathways of higher complexity . The TBC1D3 family has been identified as a hominoid-specific oncogene that can enhance cell proliferation and is encoded by a cluster of eight paralogs on chromosome 17 .
TBC1D3G antibodies are primarily validated for the following applications:
The choice of application depends on your experimental question. For protein expression analysis across different samples, Western Blot is typically used. For localization studies, ICC or IHC are preferred methods. ELISA is useful for quantitative protein detection from solution samples .
Proper control selection is critical for valid experimental interpretation when working with TBC1D3G antibodies:
Positive Controls:
Recombinant TBC1D3G protein (available as catalog numbers ABIN7544194 or ABIN3073765)
Triple-negative breast cancer (TNBC) tissue samples, which show higher expression of TBC1D3G compared to other breast cancer subtypes
Negative Controls:
Fluorescence Minus One (FMO) controls for flow cytometry experiments to determine gating boundaries
TBC1D3G knockout cell lines using CRISPR-Cas9 or siRNA approaches
Mouse tissue samples (as TBC1D3 is a hominoid-specific gene)
Avoid using isotype controls as they often have different binding properties and fluorophore-to-antibody ratios compared to your test antibody . For flow cytometry, FMO controls are more reliable for establishing positive signal thresholds.
Sample preparation significantly impacts TBC1D3G antibody performance:
When analyzing flow cytometry data with TBC1D3G antibodies, follow these methodological steps:
Gating Strategy:
Statistical Analysis:
Report percentage of positive cells for frequency analysis
Use Median Fluorescence Intensity (MFI) rather than mean for expression level analysis (as flow cytometry data is typically displayed on logarithmic scales)
Calculate fold-change in MFI as MFI(sample)/MFI(control) when comparing expression levels between samples
Visualization:
Remember that small changes in negative populations can translate into large changes in fold-difference due to the logarithmic scale, so interpret fold-changes with caution .
Several challenges exist when interpreting results from TBC1D3G antibody experiments:
Cross-reactivity: TBC1D3G belongs to a family with several paralogs (including TBC1D3, TBC1D3B, TBC1D3C) that share significant sequence homology . Verify antibody specificity against these related proteins.
Isoform specificity: Different antibodies may recognize different epitopes and potentially different TBC1D3G isoforms. For example, some antibodies target regions such as amino acids 231-281 or 312-549 .
Species considerations: TBC1D3 is a hominoid-specific gene, so results from non-primate models must be interpreted with caution . Most commercial antibodies show reactivity to human TBC1D3G .
Background signals: In immunohistochemistry, autofluorescence can be mistaken for positive staining. Always include proper unstained controls .
Quantification methods: For Western blot analysis, normalize TBC1D3G signals to appropriate loading controls (such as GAPDH) as demonstrated in previous studies .
TBC1D3G has been implicated in cancer progression and immune responses, making it an important target for advanced research:
Cancer Biomarker Studies:
TBC1D3G is expressed at higher levels in triple-negative breast cancers (TNBC) compared to other breast cancer subtypes
TBC1D3 family can be used as a prognostic biomarker in kidney renal clear cell carcinoma (KIRC)
Flow cytometry with TBC1D3G antibodies can identify cells with altered TBC1D3G expression levels in patient-derived samples
Immune Response Investigation:
TBC1D3 family expression correlates with immune checkpoint inhibitors including CD160, CTLA4, CD244, LAG3, PDCD1, and TIGIT
TBC1D3G expression is associated with CD4+ T cell infiltration levels in tumor microenvironments
Correlation analysis between TBC1D3 expression and marker sets of neutrophils, Th1, Th2, Treg, and T cell exhaustion revealed significant relationships
Methodology Approaches:
Combine TBC1D3G antibody staining with other immune cell markers for multiparameter flow cytometry
Use immunoprecipitation with TBC1D3G antibodies followed by mass spectrometry to identify interacting partners
Employ tissue microarrays with TBC1D3G antibodies to analyze expression patterns across large numbers of patient samples
TBC1D3G's function as a GTPase activating protein for RAB5 suggests its involvement in membrane trafficking pathways. Several methodological approaches can be used to investigate this role:
Colocalization Studies:
Functional Assays:
Protein-Protein Interaction Analysis:
Domain Mapping:
Recent research has identified TBC1D3G as potentially involved in antibody-dependent enhancement (ADE) of dengue virus (DENV) infection . Methodological approaches to investigate this function include:
Infection Models:
Develop ADE assays using cells expressing TBC1D3G and FcγRIIa
Compare binding and internalization of IgG-DENV complexes in control versus TBC1D3G knockout cells
Mechanistic Studies:
Use TBC1D3G antibodies for immunofluorescence to track its localization during viral entry
Investigate colocalization with FcγRIIa and viral particles during ADE
Functional Validation:
Generate TBC1D3G knockout cell lines using CRISPR-Cas9
Complement knockout cells with wild-type or mutant TBC1D3G constructs
Assess binding of IgG-virus complexes with flow cytometry
Measure virus internalization and replication in cells with varying TBC1D3G expression
Translational Application:
Screen inhibitors that disrupt TBC1D3G function to identify potential therapeutic agents against ADE
Analyze TBC1D3G expression in patients with severe versus mild dengue infections
Current TBC1D3G antibodies have several limitations that researchers should be aware of:
Specificity challenges: Due to the high sequence similarity between TBC1D3 family members, achieving absolute specificity for TBC1D3G over related proteins (TBC1D3, TBC1D3B, TBC1D3C) remains challenging .
Validation status: Many commercially available antibodies have limited validation data. Only a few antibodies have been rigorously validated in knockout systems or across multiple applications .
Application restrictions: Some antibodies are validated only for specific applications such as ELISA or Western blot but not for immunoprecipitation or flow cytometry .
Species reactivity: Most antibodies primarily target human TBC1D3G with limited cross-reactivity to other species, reflecting its hominoid-specific nature .
Epitope accessibility: Depending on experimental conditions, certain epitopes may be masked by protein folding or interactions with other molecules.
Several cutting-edge approaches could advance TBC1D3G research beyond traditional antibody methods:
CRISPR-Cas9 gene editing:
Generate endogenously tagged TBC1D3G (with GFP, mCherry, or HaloTag) to track the protein without antibodies
Create precise point mutations to study structure-function relationships
Single-cell transcriptomics:
Proximity-based labeling:
BioID or APEX2 fusion proteins to identify the TBC1D3G interactome without relying on antibody-based precipitation
Spatial mapping of TBC1D3G interactions in different cellular compartments
Structural biology approaches:
Cryo-EM or X-ray crystallography of TBC1D3G in complex with RAB5 to understand mechanistic details
Molecular dynamics simulations to predict functional consequences of mutations
Multi-omics integration:
These emerging methods could provide more comprehensive insights into TBC1D3G's biology while addressing the limitations of current antibody-based approaches.