TMEM176A antibodies are immunoglobulin-based reagents designed to bind specifically to the TMEM176A protein, which spans cellular membranes and plays roles in ion channel activity and immune modulation . Key characteristics include:
Target: Transmembrane protein 176A (UniProt accession: Q8IUR5)
Molecular weight: 26.1 kDa (235 amino acids)
Subcellular localization: Membrane-associated, particularly in the Golgi apparatus and endolysosomal compartments .
Applications: Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and flow cytometry .
TMEM176A forms cation channels with its homolog TMEM176B, influencing dendritic cell (DC) function:
Regulates antigen processing and presentation via MHC class II .
Colocalizes with HLA-DM in late endolysosomal vesicles, critical for CD4+ T cell priming .
Knockout studies in mice show impaired DC-mediated T cell activation .
Elevated TMEM176A correlates with tumor progression:
Ion Channel Function: TMEM176A/B form acid-sensitive cation channels critical for endolysosomal pH regulation, affecting antigen cross-presentation in DCs .
Epigenetic Regulation: Hypermethylation of the TMEM176A promoter silences expression, correlating with poor prognosis in colorectal cancer .
Therapeutic Target: In GBM, TMEM176A knockdown reduces ERK1/2 activation and tumor growth, while overexpression accelerates cell cycle progression .
Prognostic Marker: TMEM176A methylation status independently predicts 5-year survival in colorectal cancer (HR = 2.16, P = 0.044) .
Immune Evasion: Overexpression in hepatocellular carcinoma (HCC) suppresses antitumor immunity by altering DC function .
Validation: Antibodies like HPA008770 (Sigma-Aldrich) are validated using recombinant protein expression and tissue microarrays .
Staining Patterns: Cytoplasmic and membranous staining observed in IHC, with reduced expression in cancer versus normal tissues .
Cross-Reactivity: Most antibodies target human TMEM176A but may cross-react with mouse and rat orthologs .
TMEM176A is a transmembrane protein that has been implicated in several pathological processes, particularly in cancer and immune regulation. The protein consists of 235 amino acids with a molecular weight of approximately 26-28 kDa, and contains multiple transmembrane domains .
TMEM176A antibodies are critical research tools because:
They enable detection and quantification of TMEM176A expression in various tissues and cell types
They help elucidate TMEM176A's role in normal physiology and disease states
They facilitate the study of TMEM176A's involvement in key cellular pathways, particularly ERK signaling
They allow investigation of TMEM176A's function in dendritic cell maturation and immune regulation
Research has shown that TMEM176A expression is frequently dysregulated in certain cancers, making it a potential biomarker and therapeutic target, particularly in lung cancer and hepatocellular carcinoma .
TMEM176A antibodies can be utilized across multiple experimental platforms:
For optimal results, researchers should validate antibody performance in their specific experimental systems before conducting comprehensive studies. Antigen retrieval methods significantly impact detection sensitivity in IHC applications, with Tris-EDTA buffer (pH 9.0) generally providing better results than citrate buffer (pH 6.0) .
The selection between monoclonal and polyclonal antibodies depends on experimental goals:
Monoclonal TMEM176A Antibodies:
Provide high specificity and reproducibility
Example: Mouse monoclonal antibody against full-length recombinant TMEM176A (AA 1-235)
Ideal for applications requiring consistent lot-to-lot performance
Better for detecting specific epitopes in their native conformation
Typically used when comparing TMEM176A expression levels across multiple samples
Polyclonal TMEM176A Antibodies:
Recognize multiple epitopes on the TMEM176A protein
Examples: Rabbit polyclonal antibodies targeting different regions
Offer higher sensitivity, particularly useful in low-expression contexts
More tolerant of protein denaturation, making them versatile for various applications
Preferable for initial characterization studies or when protein structure may be altered
For methylation studies in cancer tissues, polyclonal antibodies may provide better detection across heterogeneously methylated samples, while monoclonal antibodies might offer more consistent results for quantitative analyses .
Proper controls are essential for accurately interpreting TMEM176A antibody results:
Recommended Positive Controls:
Recommended Negative Controls:
Primary antibody omission
Isotype control antibodies (IgG1 for mouse monoclonal antibodies)
Cell lines with TMEM176A knockdown
Tissue samples with confirmed TMEM176A methylation (for expression studies)
For comprehensive validation, researchers should include both technical controls (antibody specificity) and biological controls (tissues/cells with known expression patterns). When studying methylation effects, unmethylated and methylated lung cancer samples can serve as biological controls .
Researchers frequently encounter several challenges when working with TMEM176A antibodies:
Non-specific binding: Particularly problematic in tissues with high background. Solution: Optimize blocking conditions and antibody dilutions; consider using highly validated antibodies like Prestige Antibodies .
Variable epitope accessibility: TMEM176A is a multi-pass membrane protein, making some epitopes difficult to access. Solution: Test antibodies targeting different regions (N-terminal, internal, C-terminal) .
Detection in fixed tissues: Formalin fixation can mask epitopes. Solution: Implement appropriate antigen retrieval methods, particularly heat-mediated retrieval with Tris-EDTA buffer (pH 9.0) .
Methylation interference: TMEM176A promoter methylation can affect expression levels, complicating antibody-based detection. Solution: Consider complementing protein detection with methylation analysis .
Cross-reactivity with TMEM176B: TMEM176A and TMEM176B share sequence homology. Solution: Validate antibody specificity using recombinant protein controls .
TMEM176A antibodies serve as valuable tools in cancer research, particularly for:
Diagnostic biomarker development: TMEM176A methylation is found in 53.66% of primary lung cancers, making it a potential diagnostic marker . Antibodies can help correlate methylation status with protein expression.
Therapeutic target validation: Research has shown that restoration of TMEM176A expression induces cancer cell apoptosis and G2/M phase arrest while inhibiting colony formation, proliferation, migration, and invasion . Antibodies can monitor TMEM176A expression following experimental interventions.
Pathway analysis: TMEM176A suppresses tumor growth by inhibiting ERK signaling. Antibodies can be used in combination with phospho-ERK antibodies to study this regulatory relationship .
Drug sensitivity studies: Methylation of TMEM176A sensitizes cancer cells to AZD0156, an ATM inhibitor. Antibodies can help identify tumors likely to respond to such targeted therapies .
Xenograft model evaluation: TMEM176A has been shown to suppress H1299 cell xenograft growth in mice. Antibodies can monitor TMEM176A expression in these models .
For cancer research applications, combining antibody-based detection with methylation analysis provides the most comprehensive understanding of TMEM176A's role in tumorigenesis and potential therapeutic applications.
Integrating antibody detection with methylation analysis requires a multi-faceted approach:
Sequential analysis workflow:
Assess methylation status using methylation-specific PCR (MSP)
Determine protein expression using validated TMEM176A antibodies
Correlate methylation patterns with protein expression levels
Experimental design considerations:
Include cell lines with known methylation status as controls
Use demethylating agents (e.g., 5-aza-2'-deoxycytidine) to restore expression
Confirm specificity with multiple antibodies targeting different epitopes
Recommended protocols:
This integrated approach has successfully demonstrated that TMEM176A expression is regulated by promoter region methylation in lung cancer, with significant implications for understanding cancer biology and developing targeted therapies .
TMEM176A and its related protein TMEM176B are increasingly recognized as important regulators of dendritic cell maturation and immune responses:
Dendritic cell maturation studies:
Immune dysfunction in spinal cord injury (SCI):
Therapeutic development approaches:
Researchers studying neuroinflammation or neuroimmune interactions should consider incorporating TMEM176A antibodies into their experimental design, particularly when investigating dendritic cell function in the context of central nervous system injuries .
Several promising research directions for TMEM176A antibodies are emerging:
Synthetic lethality therapeutics: TMEM176A methylation status sensitizes cancer cells to ATM inhibitors like AZD0156. Antibodies could be developed as companion diagnostics to identify patients likely to respond to such targeted therapies .
Neuroprotection strategies: Given TMEM176A's role in immune regulation after spinal cord injury, antibodies could facilitate the development of immune-based therapies to promote tissue regeneration and limit further damage in chronic SCI .
Single-cell protein profiling: Advances in single-cell technologies could incorporate TMEM176A antibodies to understand cellular heterogeneity in cancer and immune responses.
Spatial transcriptomics integration: Combining TMEM176A antibody-based protein detection with spatial transcriptomics could reveal location-dependent regulation in complex tissues.
Post-translational modification studies: Developing antibodies specific to modified forms of TMEM176A could reveal regulatory mechanisms beyond methylation-controlled expression.
To rigorously test novel hypotheses about TMEM176A function, researchers should implement comprehensive validation strategies:
Multiple antibody approach:
Use antibodies targeting different TMEM176A epitopes
Compare results from different antibody classes (monoclonal vs. polyclonal)
Validate with both commercial and custom-developed antibodies
Complementary technique integration:
Combine protein detection (antibody-based) with gene expression analysis
Correlate with methylation studies in relevant contexts
Support with functional assays (e.g., proliferation, apoptosis, migration)
Model system diversity:
Test hypotheses across multiple cell lines
Validate in primary patient samples
Confirm in appropriate animal models
Pathway analysis validation:
Assess effects on known interacting pathways (particularly ERK signaling)
Perform co-immunoprecipitation to identify binding partners
Utilize inducible expression systems to study temporal dynamics