TBC1D14 antibodies target the TBC1D14 protein, a Rab GTPase-activating protein (RabGAP) that regulates autophagy by modulating interactions between recycling endosomes (REs), the TRAPP complex, and ATG9 vesicles . TBC1D14 acts as a negative regulator of autophagosome formation and plays roles in secretory traffic and cancer metastasis .
Multiple antibodies have been developed and validated for TBC1D14 detection. Key examples include:
These antibodies are widely used in Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) .
TBC1D14 inhibits autophagy by binding to the TRAPPIII complex, which regulates RAB1 activation and ATG9 vesicle trafficking .
Overexpression of TBC1D14 disrupts Golgi function and secretory traffic by interacting with TRAPPC8, a TRAPPIII subunit .
In head and neck squamous cell carcinoma (HNSCC), TBC1D14 suppresses lymph node metastasis (LNM) by inhibiting autophagy. Low TBC1D14 expression correlates with poor prognosis .
Mechanistically, TBC1D14 downregulates MAEA (macrophage erythroblast attacher), a protein that promotes autophagy and metastasis .
TBC1D14 binds ULK1 (a key autophagy kinase) and RAB11, linking RE dynamics to autophagosome formation .
Proteomic analyses identified TRAPP complex subunits and MAEA as critical interactors, highlighting its dual role in membrane trafficking and tumor suppression .
Western Blotting: Detects endogenous TBC1D14 at ~100 kDa in HEK293A and HNSCC cell lines .
Immunohistochemistry: Used to quantify TBC1D14 expression in HNSCC patient tissues, revealing its negative correlation with MAEA .
Functional Studies: siRNA knockdown and overexpression models demonstrate TBC1D14's role in autophagy and metastasis .
TBC1D14 (TBC1 Domain Family, Member 14) is a protein that functions as a negative regulator of autophagy. It acts primarily by controlling membrane delivery from RAB11-positive recycling endosomes to forming autophagosomes . TBC1D14 contains a TBC domain at its C-terminus (residues 411-611), which is characteristic of proteins with potential GTPase-activating protein (GAP) activity for Rab family proteins .
The protein has been shown to interact with the TRAPP (trafficking protein particle) complex via its N-terminal region (amino acids 120-223), also known as the TRAPP-binding region (TBR) . This interaction is crucial for regulating both autophagic and secretory pathways, as TBC1D14 appears to function as a bridge between a TRAPP complex and activated RAB11 . Importantly, while TBC1D14 can bind to RAB11, it may not act as a GAP for RAB11 but rather as an effector .
Beyond its role in autophagy, recent research has implicated TBC1D14 in cancer progression. Studies have shown that TBC1D14 has lower expression in head and neck squamous cell carcinoma (HNSCC) with lymph node metastasis and may serve as a favorable prognostic indicator .
Commercial TBC1D14 antibodies target various regions of the protein, with significant functional and experimental implications. The choice of epitope can dramatically affect experimental outcomes based on the protein domains and interactions being studied.
When selecting a TBC1D14 antibody, researchers should consider which functional domain they're investigating. For studying TRAPP complex interactions, antibodies targeting the N-terminal region may be more informative, while those investigating potential GAP activity might prefer antibodies targeting the C-terminal TBC domain .
Validating TBC1D14 antibody specificity is crucial for ensuring reliable experimental results. A comprehensive validation approach should include:
Western blot analysis with positive and negative controls:
Immunoprecipitation validation:
Immunofluorescence and colocalization:
Genetic approaches:
Use siRNA knockdown of TBC1D14 and show reduced antibody signal
For advanced validation, employ CRISPR-Cas9 knockout cells as negative controls
Studying TBC1D14's role in autophagy regulation requires sophisticated approaches leveraging specific antibodies:
Monitoring autophagy inhibition:
Visualizing recycling endosome-autophagosome interactions:
Perform triple immunofluorescence using antibodies against TBC1D14, RAB11 (recycling endosome marker), and autophagy proteins (ULK1, ATG9, LC3)
This approach reveals how TBC1D14 mediates membrane trafficking from recycling endosomes to forming autophagosomes
Time-lapse imaging with fluorescently tagged proteins can complement antibody-based fixed-cell approaches
Analyzing TBC1D14-TRAPP complex interactions during autophagy:
Proximity labeling approaches:
Analysis of TBC1D14-TRAPP interactions requires tailored methodological approaches:
Co-immunoprecipitation (Co-IP) protocol:
Lyse cells in buffer containing 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, and protease inhibitors
Incubate cell lysate with TBC1D14 antibody (e.g., ABIN955090) overnight at 4°C
Add protein A/G beads and incubate for 2-3 hours
Wash with lysis buffer and elute with SDS sample buffer
Analyze by immunoblotting for TRAPP complex subunits (TRAPPC4, TRAPPC8, TRAPPC12)
GST pull-down assay:
Proximity labeling with BioID:
Immunofluorescence co-localization:
TBC1D14's function is determined by distinct domains with unique roles that can be studied using domain-specific antibodies:
N-terminal TRAPP-binding region (TBR, aa 120-223):
ULK1-binding region (aa 224-330):
TBC domain (aa 411-611):
When using domain-specific antibodies, researchers should consider:
Domain | Function | Experimental Applications | Antibody Considerations |
---|---|---|---|
TRAPP-binding region (120-223) | TRAPP complex interaction | Track TRAPP binding, secretory pathway modulation | May interfere with TRAPP interaction |
ULK1-binding region (224-330) | ULK1 kinase interaction | Monitor autophagy initiation | May block ULK1 binding |
TBC domain (411-611) | Potential GAP activity | Study RAB regulation | Less likely to interfere with protein interactions |
Recent research has implicated TBC1D14 in cancer progression, particularly in head and neck squamous cell carcinoma (HNSCC). When studying this aspect, researchers should consider:
Expression analysis in clinical samples:
Use TBC1D14 antibodies for immunohistochemistry on HNSCC tissue microarrays
Compare expression between primary tumors with and without lymph node metastasis
Quantify expression using digital pathology algorithms
Previous studies have shown lower TBC1D14 expression in HNSCC with lymph node metastasis
Functional studies in HNSCC cell lines:
In vivo metastasis models:
Develop TBC1D14-modulated HNSCC cell lines
Implant in appropriate animal models
Monitor lymph node metastasis formation
Perform immunohistochemistry on primary tumors and metastases
Correlation with autophagy markers:
TBC1D14 overexpression generates distinctive tubulated endosomal structures that can be visualized using optimized immunofluorescence techniques:
Sample preparation protocol:
Transfect cells with GFP-TBC1D14 (or use endogenous TBC1D14 with appropriate antibodies)
Fix with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1% Triton X-100 for 5 minutes
Block with 5% normal serum in PBS for 1 hour
Antibody combinations for optimal visualization:
Primary antibody set 1: TBC1D14 + RAB11 + RAB1B
Primary antibody set 2: TBC1D14 + TRAPPC4 + RAB11
Primary antibody set 3: TBC1D14 + TRAPPC4 + RAB1B
Advanced imaging considerations:
Use confocal microscopy with high NA objectives (≥1.3) for optimal resolution
Consider super-resolution approaches (STED, SIM, STORM) for detailed tubule morphology
Perform live-cell imaging with fluorescently tagged proteins to observe tubule dynamics
Take Z-stacks (0.3μm steps) to capture the full 3D structure of tubules
Quantitative analysis:
Measure tubule length, number per cell, and branching pattern
Quantify co-localization coefficients between TBC1D14 and various markers
Compare tubulation under different conditions (starvation, drug treatments)
Rigorous experimental controls are critical for generating reliable data with TBC1D14 antibodies:
Western blotting controls:
Positive control: Lysate from cells with confirmed TBC1D14 expression
Negative control: Lysate from TBC1D14 knockdown/knockout cells
Loading control: Housekeeping protein (e.g., β-actin, GAPDH)
Secondary antibody-only control: Check for non-specific binding
Immunoprecipitation controls:
IgG control: Non-specific IgG from the same species as the TBC1D14 antibody
Input control: Analyze a portion of the lysate before IP
Specificity control: Perform IP with and without TBC1D14 overexpression
Immunofluorescence controls:
Primary antibody omission: Detect secondary antibody non-specific binding
Blocking peptide competition: Confirm epitope specificity
Knockdown validation: Reduced signal in TBC1D14-depleted cells
For tubulation experiments: Compare with RAB11 staining patterns
Function-blocking experiments:
When using antibodies to disrupt TBC1D14 function, include:
Isotype control antibodies
Dose-response curves
Timing controls (pre-incubation, co-incubation)
Detecting endogenous TBC1D14 presents several challenges that can be addressed with specialized approaches:
Low expression levels:
Challenge: TBC1D14 may be expressed at low levels in some cell types
Solution: Use signal amplification methods like TSA (tyramide signal amplification) for immunofluorescence
Alternative: Concentrate proteins by immunoprecipitation before Western blotting
Antibody specificity issues:
Challenge: Cross-reactivity with related TBC domain proteins
Solution: Validate with genetic approaches (siRNA, CRISPR knockout)
Alternative: Use multiple antibodies targeting different epitopes
Subcellular localization:
Challenge: Detecting dispersed protein across recycling endosomes
Solution: Use super-resolution microscopy techniques
Alternative: Employ subcellular fractionation followed by Western blotting
Dynamic regulation during autophagy:
Challenge: Capturing temporal changes in localization
Solution: Use synchronized cell populations with timed autophagy induction
Alternative: Live-cell imaging with knock-in fluorescent tags (CRISPR knock-in)
Combining TBC1D14 antibodies with proximity labeling offers powerful insights into its interaction network:
BioID approach optimization:
Generate myc-BioID-TBC1D14 fusion constructs
Express in target cells and provide biotin (50 μM) for 24 hours
Perform streptavidin pull-downs under denaturing conditions (1% SDS)
Analyze bound proteins by mass spectrometry
Previous studies identified TRAPPC8 as most proximal to TBC1D14 using this approach
APEX2 proximity labeling:
Create APEX2-TBC1D14 fusion proteins
Optimize H₂O₂ treatment (1 mM, 1 minute) for biotinylation
Capture labeled proteins under various conditions (basal, starvation)
Compare interactomes during different cellular states
Verification with conventional approaches:
Confirm key interactions using TBC1D14 antibodies for co-immunoprecipitation
Perform reciprocal IPs with antibodies against identified partners
Use immunofluorescence to confirm co-localization of TBC1D14 and partners
Domain-specific interactome analysis:
Create truncation constructs (TBR domain, ULK1-binding region, TBC domain)
Perform domain-specific proximity labeling
Map interaction sites for different partners
Cross-validate with in vitro binding assays
Issue | Possible Causes | Solutions |
---|---|---|
No signal in Western blot | Low expression levels, Antibody degradation, Incorrect epitope accessibility | Increase protein loading, Use fresh antibody, Try different extraction buffers, Consider different antibody |
Multiple bands | Cross-reactivity, Protein degradation, Post-translational modifications | Validate with siRNA knockdown, Add protease inhibitors, Consider phosphatase treatment |
No co-immunoprecipitation of known partners | Harsh lysis conditions, Epitope masking, Transient interactions | Use milder detergents, Try different antibodies, Consider crosslinking before lysis |
No tubulation in GFP-TBC1D14 overexpression | Insufficient expression, Cell type differences, Mutated construct | Optimize transfection, Try different cell lines, Sequence verify construct |
Background in immunofluorescence | Non-specific binding, Autofluorescence, Fixation artifacts | Optimize blocking, Use Sudan Black to reduce autofluorescence, Test different fixation methods |
Resolving inconsistencies across experimental systems requires systematic troubleshooting:
Cell type-specific differences:
TBC1D14 function may vary between cell types due to different expression levels of interaction partners
Solution: Characterize the expression of key interactors (RAB11, TRAPP components) in each cell system
Validate findings across multiple cell lines representing the tissue of interest
Antibody performance variation:
Different antibodies may recognize distinct epitopes/conformations
Solution: Test multiple validated antibodies targeting different regions
Consider creating a consensus from multiple antibody results
Technical variability:
Fixation methods can affect epitope accessibility in immunofluorescence
Solution: Standardize protocols across laboratories
Include positive controls with known outcomes in each experiment
Functional redundancy:
Other TBC domain proteins may compensate for TBC1D14
Solution: Consider double knockdown experiments
Analyze expression of related proteins in different systems