TMEM127 antibody is a polyclonal rabbit immunoglobulin (IgG) designed to detect the transmembrane protein 127 (TMEM127), a tumor suppressor implicated in pheochromocytomas, paragangliomas, and renal carcinomas. TMEM127 regulates membrane organization, endosomal trafficking, and mTOR signaling, making it a critical target in oncology research. The antibody is used in Western blot, immunofluorescence, immunoprecipitation (IP), and immunohistochemistry (IHC) to study protein localization, expression levels, and functional interactions .
Immunogen: Synthetic peptide targeting the middle region of human TMEM127 (e.g., sequence: AFLLDVFGPKHPALKITRRYAFAHILTVLQCATVIGFSYWASELILAQQQ) .
Species Reactivity: Primarily human; cross-reactivity with mouse in some formulations .
Plasma Membrane Disorganization: Loss of TMEM127 disrupts lipid domains, impairing clathrin-mediated endocytosis (CME) and causing surface accumulation of receptor tyrosine kinases (RTKs) like RET .
RET Signaling: TMEM127 deficiency increases cell surface RET, leading to ligand-independent activation and sustained proliferation in pheochromocytoma (PCC) models .
Therapeutic Implications: RET inhibitors (e.g., selpercatinib) reduce tumor growth in TMEM127-mutant PCC xenografts .
mTORC1 Regulation: TMEM127 interacts with the Rag-GTPase/LAMTOR complex, reducing mTORC1 recruitment to lysosomes under nutrient-rich conditions .
Lysosomal Acidification: TMEM127 binds vATPase in an acid-dependent manner, modulating LAMTOR-vATPase associations .
Insulin Sensitivity: TMEM127 knockout mice exhibit enhanced insulin receptor signaling and AKT activation .
Immune Modulation: TMEM127 depletion increases MHC-II surface expression in infected cells, altering T-cell responses .
| Supplier | Catalog Number | Host | Applications | Immunogen | Species Reactivity |
|---|---|---|---|---|---|
| Novus Biologicals | NBP1-91305 | Rabbit | Western Blot | Synthetic peptide (middle region) | Human |
| Proteintech | 23142-1-AP | Rabbit | WB, IHC, IF, IP, ELISA | Full-length TMEM127 fusion protein | Human, Mouse |
| Sigma-Aldrich | SAB2102452 | Rabbit | Western Blot | Synthetic peptide (unspecified region) | Human, Mouse, Others |
TMEM127 is a tumor suppressor gene encoding a transmembrane protein that is frequently mutated in pheochromocytomas and, less commonly, in renal cancers. The significance of TMEM127 in research stems from its role in regulating mTORC1 signaling through interaction with the lysosome-anchored complex comprising Rag GTPases, the LAMTOR pentamer, and vATPase . This complex plays a crucial role in amino acid-mediated mTORC1 activation. Mutations in TMEM127 lead to increased mTORC1 signaling, contributing to tumorigenesis through mechanisms that researchers are actively investigating . Understanding TMEM127's function is essential for developing targeted therapies for cancers with TMEM127 mutations.
TMEM127 antibodies can be used in multiple experimental applications, including:
| Application | Recommended Dilution | Notes |
|---|---|---|
| Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate | Validated in HeLa cells |
| Immunohistochemistry (IHC) | 1:250-1:1000 | Validated in human stomach cancer tissue, heart tissue, and liver cancer tissue |
| Immunofluorescence (IF)/ICC | 1:200-1:800 | Validated in NIH/3T3 cells |
| Western Blot (WB) | Refer to published literature | Documented in multiple publications |
| ELISA | Optimization required | Sample-dependent |
For optimal results, titration of the antibody is recommended for each experimental system .
Validation of TMEM127 antibody specificity should include:
Negative controls using TMEM127 knockout cells, as demonstrated in studies with HEK293T TMEM127-KO cells .
Positive controls using tissues known to express TMEM127, such as heart tissue or specific cancer tissues .
Peptide competition assays to confirm binding specificity.
Multiple detection methods (e.g., Western blot and immunofluorescence) to confirm consistent results.
Comparison with recombinant TMEM127 expression systems, particularly when studying specific variants.
When working with suspected TMEM127 mutations, researchers should compare antibody reactivity between wild-type and mutant TMEM127 to assess potential epitope alterations .
For optimal immunodetection of TMEM127:
Immunohistochemistry (IHC):
Antigen retrieval: Use TE buffer pH 9.0 (primary recommendation) or citrate buffer pH 6.0 (alternative)
Working dilution: 1:250-1:1000
Positive control tissues: Human stomach cancer tissue, heart tissue, liver cancer tissue
Immunofluorescence:
Fixation: 4% paraformaldehyde in PBS is suitable for most applications
Permeabilization: Different permeabilization agents reveal different aspects of TMEM127 localization
Immunoprecipitation:
Lysate preparation: Use 1.0-3.0 mg of total protein lysate
TMEM127 localizes to multiple subcellular compartments, complicating its detection and analysis. To differentiate between various localizations:
Co-localization studies: Use confocal microscopy with markers for:
Lysosomes (LAMP1, LAMP2)
Early endosomes
Late endosomes
Plasma membrane
Membrane fractionation: Separate cytosolic and membrane-enriched fractions. TMEM127 is predominantly detected in membrane-enriched fractions containing lysosomes (LAMP2 positive) but not in cytosolic fractions .
Time-course experiments: After amino acid stimulation, TMEM127 colocalization with lysosomal markers (LAMP2) increases, peaking at approximately 10 minutes before gradually decreasing . This dynamic profile resembles mTORC1 association with LAMP2.
Permeabilization techniques: Use selective membrane permeabilization to distinguish between plasma membrane and internal membrane localization:
Determining TMEM127 membrane topology requires multiple complementary approaches:
Antibody accessibility assays: Using antibodies directed against different termini of TMEM127 under various permeabilization conditions. Research has shown that both N- and C-termini of TMEM127 are oriented toward the cytoplasm, supporting an even number of transmembrane domains .
Computational prediction tools: Hydrophobicity analyses and transmembrane domain prediction algorithms can identify potential membrane-spanning regions. These predictions should be experimentally validated .
Terminal tagging strategies: Creating constructs with epitope tags at either terminus allows for orientation determination using selective permeabilization.
Protease protection assays: Regions exposed to the cytoplasm are susceptible to protease digestion, while lumenal domains are protected.
Recent research has revealed that TMEM127 contains four transmembrane domains, contradicting earlier models suggesting only three domains. A previously unrecognized transmembrane domain was identified between residues 30 and 53 .
To analyze TMEM127 interaction with mTORC1 signaling:
Co-immunoprecipitation studies:
Amino acid stimulation experiments:
Rag GTPase binding assays:
Lysosomal recruitment analysis:
Functional characterization of TMEM127 variants requires multiple methodological approaches:
Subcellular localization analysis:
Protein stability assessment:
Membrane association studies:
Functional rescue experiments:
Recent studies have identified three subgroups of mutations and determined that approximately 71% of tumor-associated variants, including 60% of missense variants, are pathogenic or likely pathogenic due to loss of membrane binding ability, stability, and/or internalization capability .
TMEM127 undergoes internalization through clathrin-mediated endocytosis, providing a model for studying this process:
Internalization assays:
Label cell surface TMEM127 using antibodies against extracellular domains or epitope tags
Allow internalization at 37°C for various time points
Remove remaining surface label and quantify internalized protein
Mutational analysis of trafficking motifs:
Colocalization with endocytic markers:
Track TMEM127 progression through early endosomes, late endosomes, and lysosomes
Use markers such as EEA1 (early endosomes), Rab7 (late endosomes), and LAMP1/2 (lysosomes)
Endocytic pathway inhibitors:
Use chlorpromazine or dynasore to inhibit clathrin-mediated endocytosis
Apply filipin or methyl-β-cyclodextrin to disrupt caveolae-mediated endocytosis
Determine which pathway predominantly regulates TMEM127 trafficking
When facing conflicting TMEM127 localization data:
Consider amino acid status: TMEM127 localization is dynamic and dependent on amino acid availability. Colocalization with lysosomal markers increases after amino acid exposure, peaking around 10 minutes before gradually decreasing .
Evaluate expression levels: Overexpression can lead to saturation of trafficking machinery and mislocalization. Compare endogenous localization with various expression levels of recombinant protein.
Cell type variations: TMEM127 exhibits ubiquitous expression but may show cell type-specific localization patterns. Compare findings across multiple cell lines.
Antibody epitope accessibility: TMEM127's complex membrane topology may result in epitope masking in certain cellular compartments. Use multiple antibodies targeting different regions of the protein.
Fixation and permeabilization effects: Different methods can selectively preserve or expose certain localizations:
When analyzing TMEM127-null tumors or cell models:
Validation of TMEM127 loss: Confirm complete absence of TMEM127 expression using multiple methods (Western blot, qPCR, immunostaining).
Expected molecular changes:
Functional consequences:
Compensatory mechanisms: Long-term TMEM127 loss may activate compensatory pathways that mask acute effects.
Genetic background considerations: The impact of TMEM127 loss may vary depending on the status of other genes in the mTORC1 pathway.
Common pitfalls in TMEM127 immunoprecipitation experiments include:
Epitope masking: TMEM127's membrane association and protein interactions may obscure antibody binding sites. Test multiple antibodies targeting different regions.
Cross-reactivity: Validate antibody specificity using TMEM127-knockout controls to ensure signals are specific .
Detergent selection: Membrane protein extraction requires careful detergent selection:
Too harsh: May disrupt protein-protein interactions
Too mild: May fail to solubilize membrane-bound TMEM127
Recommended: CHAPS, NP-40, or Triton X-100 at optimized concentrations
Transient interactions: Some TMEM127 interactions (e.g., with LAMTOR components) are dynamic and amino acid-dependent . Consider crosslinking or stimulus-specific timing.
Low expression levels: Endogenous TMEM127 is often expressed at low levels. Scale up input material or use enrichment strategies.
Sample processing: Rapid processing is essential as TMEM127-LAMTOR interactions may be unstable during prolonged handling.
TMEM127 antibodies can advance understanding of pheochromocytoma pathogenesis through:
Tumor sample analysis:
Primary pheochromocytomas carrying germline truncating TMEM127 mutations show loss of the wild-type allele and no residual TMEM127 expression
These tumors exhibit consistently higher levels of LAMTOR1 and LAMTOR2 proteins compared to pheochromocytomas with wild-type TMEM127
Trends toward increased Rag and vATPase protein levels suggest broader effects on lysosomal scaffold components
Diagnostic applications:
Immunohistochemical screening of tumor samples for TMEM127 expression
Correlation of TMEM127 loss with mTORC1 pathway activation markers
Identification of potential therapeutic vulnerabilities
Mechanistic insights:
Analysis of mTORC1 signaling components in TMEM127-mutant vs. wild-type tumors
Examination of lysosomal function and nutrient sensing in tumor samples
Investigation of potential metabolic adaptations associated with TMEM127 loss
Comparative studies:
Compare TMEM127-mutant pheochromocytomas with other genetic subtypes
Assess overlap with tumors carrying mutations in other mTORC1 pathway components
Identify molecular subgroups that might benefit from specific therapeutic strategies
To determine the functional impact of novel TMEM127 variants:
Structure-function classification:
Multi-tiered experimental assessment:
Computational prediction integration:
Functional complementation:
This multi-faceted approach has successfully classified 71% of tumor-associated variants, including 60% of missense variants, as pathogenic or likely pathogenic through loss of membrane binding ability, stability, and/or internalization capability .
When interpreting TMEM127 staining patterns in tumor samples:
Expression level assessment:
Subcellular distribution patterns:
Correlation with downstream markers:
Comparative analysis:
Compare patterns across different tumor types (pheochromocytoma, paraganglioma, renal carcinoma)
Assess correlation with genetic data when available
Consider tissue-specific differences in TMEM127 expression and localization
Controls and validation:
Include known TMEM127-mutant samples as references
Use multiple antibodies targeting different epitopes
Perform genetic analysis when unusual patterns are observed
Antibodies against TMEM127 can facilitate research into therapeutic targeting of the mTORC1 pathway through:
Biomarker development:
Identification of patient populations with TMEM127 alterations
Correlation of TMEM127 status with response to mTORC1 inhibitors
Detection of compensatory mechanisms during treatment
Mechanistic studies:
Drug development support:
Screening for compounds that modulate TMEM127-dependent mTORC1 regulation
Evaluation of combination therapies targeting both TMEM127 and mTORC1 pathways
Assessment of specificity for TMEM127-deficient vs. wild-type cells
Therapeutic resistance mechanisms:
Study of adaptive responses to mTORC1 inhibition in TMEM127-deficient contexts
Identification of bypass pathways that emerge during treatment
Development of strategies to overcome resistance
To resolve contradictions in TMEM127 membrane topology models:
Integrated structural biology approaches:
Cryogenic electron microscopy of purified TMEM127
X-ray crystallography of individual domains or full-length protein
NMR spectroscopy of specific transmembrane segments
Systematic mutagenesis:
Creation of a comprehensive panel of mutations throughout the protein
Assessment of each mutation's effect on membrane integration
Identification of critical residues for membrane topology
Advanced topology mapping:
APEX2 proximity labeling at various positions within TMEM127
Glycosylation site insertion analysis
Cysteine accessibility methods
Computational prediction refinement:
Integration of machine learning approaches with experimental data
Molecular dynamics simulations of membrane insertion and folding
Evolutionary analysis of conservation patterns in transmembrane regions
Recent research has challenged the previous three-transmembrane domain model, providing evidence for a four-transmembrane domain structure with both N- and C-termini oriented toward the cytoplasm . The newly identified transmembrane domain is located between residues 30 and 53 .
To effectively analyze TMEM127-LAMTOR interaction dynamics:
Live-cell imaging approaches:
Fluorescently tagged TMEM127 and LAMTOR components
FRET or BRET assays to monitor protein-protein interactions in real-time
Photobleaching techniques (FRAP, FLIP) to assess mobility and exchange rates
Stimulus-dependent interaction analysis:
Time-course experiments following amino acid stimulation
Selective inhibition of specific signaling components
Comparison between wild-type and mutant proteins
Proximity labeling techniques:
BioID or APEX2 fusion proteins to identify neighboring proteins
Time-resolved proximity labeling to capture dynamic interactions
Comparison of interactomes under different nutrient conditions
Structural characterization:
Cryo-EM analysis of the TMEM127-LAMTOR complex
Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces
Cross-linking mass spectrometry to identify contact points
Domain mapping:
Expression of truncation mutants to identify interaction domains
Point mutations at conserved residues
Competition assays with peptides derived from interaction interfaces
Research has shown that TMEM127 interacts with LAMTOR in an amino acid-dependent manner and decreases the LAMTOR1-vATPase association, while TMEM127-vATPase binding requires intact lysosomal acidification but is amino acid independent .