TMEM131L is a transmembrane protein implicated in intracellular protein trafficking and disease processes. Key findings include:
The HRP-conjugated TMEM131L antibody enables target detection through enzymatic reactions.
Expression Analysis: TMEM131L is significantly upregulated in glioma tissues (AUC = 0.858 for GBM/LGG diagnosis) .
Survival Correlation:
Oxidative Stress: Coexpressed with SYT1, CREB3L3, and PDX1, influencing glioma progression .
Immune Modulation: Correlates with immune checkpoint markers (e.g., PD-1, CTLA-4) and tumor-infiltrating immune cells (e.g., macrophages, dendritic cells) .
Lyophilization: Improves HRP-antibody conjugation efficiency by reducing reaction volume, yielding conjugates with 20x higher sensitivity in ELISA (1:5,000 vs. 1:25 dilution) .
Activity Retention: UV-spectroscopy and SDS-PAGE confirm preserved HRP activity post-conjugation .
TMEM131L (also known as KIAA0922) is a transmembrane protein that belongs to the TMEM131 family. Based on current research, TMEM131L functions as a membrane-associated protein that antagonizes canonical Wnt signaling by triggering lysosome-dependent degradation of Wnt-activated LRP6 . It has been demonstrated to regulate thymocyte proliferation and plays an important role in several cellular processes . Recent studies have also identified TMEM131L as a potential prognostic marker in glioma, with significant expression in glioma tissues correlating with poor patient outcomes .
The TMEM131 family proteins, including TMEM131L, contain bacterial PapD chaperone-like domains in their amino termini, which are involved in recruiting premature collagen monomers for proper assembly and secretion . The carboxy termini interact with TRAPPC8, a component of the TRAPP tethering complex, to facilitate collagen cargo trafficking from the endoplasmic reticulum to the Golgi apparatus .
The TMEM131L antibody with HRP conjugation is typically a polyclonal antibody produced in rabbit hosts. Its specifications include:
| Parameter | Specification |
|---|---|
| Category | Antibodies |
| Aliases | Transmembrane protein 131-like, TMEM131L, KIAA0922 |
| Product Type | Polyclonal Antibody |
| UniprotID | A2VDJ0 |
| Immunogen Species | Homo sapiens (Human) |
| Immunogen | Peptide sequence from Human Transmembrane protein 131-like protein (963-978AA) |
| Host Species | Rabbit |
| Species Reactivity | Human |
| Primary Application | ELISA |
| Clonality | Polyclonal |
| Isotype | IgG |
| Conjugate | HRP (Horseradish peroxidase) |
| Buffer | Preservative: 0.03% Proclin 300, Constituents: 50% Glycerol, 0.01M PBS, pH 7.4 |
| Form | Liquid |
| Storage | -20°C or -80°C; avoid repeated freezing and thawing |
Information sourced from product data sheets
HRP (Horseradish peroxidase) conjugated antibodies offer several methodological advantages in research applications:
Enhanced signal amplification: HRP conjugation significantly amplifies detection signals, increasing assay sensitivity for detecting low abundance proteins like TMEM131L .
Reduced experimental steps: Direct conjugation eliminates the need for secondary antibody incubation steps, streamlining experimental protocols and reducing potential sources of variability .
Lower background noise: HRP-conjugated primary antibodies can provide higher specificity with lower background compared to two-antibody detection systems, particularly valuable when investigating novel proteins like TMEM131L with limited characterization .
Versatile detection options: HRP conjugates can be detected using multiple substrates (chemiluminescent, colorimetric, or fluorescent), allowing researchers to optimize visualization based on equipment availability and experimental needs .
This conjugation is particularly valuable when investigating proteins expressed at low levels or when sample volume is limited, as is often the case in early TMEM131L research .
For optimal ELISA performance with HRP-conjugated TMEM131L antibody, consider the following methodological guidelines:
Antigen coating concentration: For recombinant TMEM131L protein, use 1-10 μg/ml in carbonate buffer (pH 9.6) or PBS (pH 7.4) for coating microplate wells.
Blocking solution: Use 3-5% BSA or 5% non-fat dry milk in PBS with 0.05% Tween-20 for 1-2 hours at room temperature to minimize non-specific binding.
Antibody dilution: The recommended dilution range for TMEM131L antibody, HRP conjugated is typically 1:20 to 1:50, though optimal concentration should be determined empirically for each lot .
Incubation conditions: Incubate the primary HRP-conjugated antibody for 1-2 hours at room temperature or overnight at 4°C for maximum sensitivity.
Washing protocol: Perform 4-5 washes with PBS containing 0.05% Tween-20 between steps to reduce background.
Substrate selection: TMB (3,3',5,5'-Tetramethylbenzidine) is recommended for HRP detection, with reaction stopped using 2N H₂SO₄ after appropriate color development (typically 5-30 minutes).
Controls: Include positive controls (known TMEM131L-expressing samples), negative controls (samples lacking TMEM131L), and blank wells (no primary antibody) to validate results.
Signal measurement: Read absorbance at 450nm with 620nm as reference wavelength.
When troubleshooting, ensure that the antigen is properly immobilized and accessible, as TMEM131L's transmembrane nature may affect epitope presentation.
While the primary documented application for TMEM131L antibody with HRP conjugation is ELISA , researchers can adapt it for western blotting with the following optimization strategies:
Sample preparation: Use RIPA buffer supplemented with protease inhibitors for efficient extraction of membrane-associated TMEM131L. Consider using phosphatase inhibitors if investigating phosphorylation states.
Protein loading: Load 20-50 μg of total protein per lane for cell lysates or 10-25 μg for tissue extracts.
Gel selection: Use 8-10% SDS-PAGE gels for optimal resolution of TMEM131L, which has a molecular weight of approximately 131 kDa.
Transfer conditions: Transfer to PVDF membrane (preferred over nitrocellulose for transmembrane proteins) at 30V overnight at 4°C for complete transfer of high molecular weight proteins.
Blocking optimization: Test both 5% non-fat dry milk and 3-5% BSA in TBST to determine optimal blocking for your specific application.
Antibody dilution: Begin with a 1:20 to 1:50 dilution in blocking buffer, adjusting based on signal-to-noise ratio .
Incubation parameters: Incubate with primary antibody overnight at 4°C with gentle agitation.
Stringent washing: Perform 5-6 washes with TBST, 5 minutes each, to reduce background associated with HRP-conjugated antibodies.
Detection system: Use enhanced chemiluminescence (ECL) substrates, with standard sensitivity for abundant samples or high sensitivity variants for low expression levels.
Exposure optimization: Begin with short exposures (30 seconds) and increase as needed to optimize signal detection without saturation.
Include positive controls such as cell lines known to express TMEM131L (e.g., U2OS bone osteosarcoma cells) to validate antibody specificity.
Proper validation of TMEM131L antibody specificity is critical for generating reliable research data. Implement the following control strategy:
Positive expression controls: Include samples from tissues/cells with known TMEM131L expression, such as:
Negative expression controls:
Technical controls:
Secondary antibody-only control (for two-step detection methods)
Substrate-only control to assess endogenous peroxidase activity
Isotype control antibody (rabbit IgG) to identify non-specific binding
Orthogonal validation:
Correlation with mRNA expression levels via RT-PCR
Comparison with alternative antibodies targeting different TMEM131L epitopes
Recombinant expression of tagged TMEM131L followed by parallel detection
Knockout/knockdown validation:
These rigorous controls will help distinguish specific signals from artifacts and validate the TMEM131L antibody's performance across different experimental contexts.
TMEM131L and related family proteins play critical roles in collagen secretion through their PapD chaperone-like domains and interaction with trafficking machinery. Researchers can employ HRP-conjugated TMEM131L antibody to investigate these pathways through:
Co-immunoprecipitation assays: Use the antibody to pull down TMEM131L protein complexes, followed by western blotting or mass spectrometry to identify interacting partners, particularly:
Immunofluorescence co-localization: Though not directly using the HRP conjugate (which is optimized for enzymatic detection), the same antibody clone can be used in immunofluorescence to:
Visualize co-localization with ER and Golgi markers
Track TMEM131L distribution during collagen trafficking
Assess changes in localization upon disruption of trafficking pathways
ELISA-based interaction studies: Using the HRP-conjugated antibody to:
Detect TMEM131L binding to immobilized collagen proteins
Quantify TMEM131L-collagen complex formation under various conditions
Screen for compounds that modulate TMEM131L-collagen interactions
Functional rescue experiments: In cells with TMEM131L knockdown (which show decreased extracellular type I collagen fibers) , measure restoration of collagen secretion after reconstitution with:
Full-length TMEM131L
TMEM131L lacking the PapD-L domain
TMEM131L with mutations in the C-terminal region that interacts with TRAPPC8
These approaches can help delineate TMEM131L's precise role in the evolutionarily conserved collagen trafficking pathway from ER to Golgi and beyond.
Recent research has identified TMEM131L as a potential prognostic marker in glioma . Researchers can leverage HRP-conjugated TMEM131L antibodies to further investigate this relationship through:
Tissue microarray (TMA) analysis: Apply immunohistochemistry (IHC) with HRP-conjugated TMEM131L antibody to:
Quantify TMEM131L expression across large cohorts of glioma patient samples
Correlate expression patterns with clinicopathological features like:
Histological type (GBM vs. LGG)
IDH mutation status
1p/19q codeletion status
Patient age and primary therapy outcomes
Develop standardized scoring systems for TMEM131L expression
Multiplex IHC approaches: Combine TMEM131L detection with markers for:
Survival analysis methodologies:
Kaplan-Meier survival analysis comparing patients with high vs. low TMEM131L expression
Cox regression models incorporating TMEM131L expression with other clinical variables
Development of nomogram prediction models including TMEM131L status
Functional validation in patient-derived models:
Correlate TMEM131L protein levels with invasiveness in patient-derived glioma cell lines
Assess impact of TMEM131L knockdown on glioma cell proliferation, migration, and therapy resistance
Investigate potential mechanisms connecting TMEM131L to poor prognosis
The diagnostic efficacy of TMEM131L for glioma appears promising with reported area under curve (AUC) values of 0.858 and confidence interval (CI) of 0.841–0.874 for GBM and LGG , warranting further exploration of its clinical utility.
TMEM131L has been implicated in antagonizing canonical Wnt signaling by triggering lysosome-dependent degradation of Wnt-activated LRP6 . To explore this relationship, researchers can employ the following methodological approaches with HRP-conjugated TMEM131L antibody:
Biochemical interaction studies:
Co-immunoprecipitation followed by western blotting to detect associations between TMEM131L and:
LRP6 (particularly phosphorylated forms)
Other Wnt pathway components (β-catenin, GSK3β, Axin)
Lysosomal proteins involved in LRP6 degradation
ELISA-based quantification of TMEM131L-LRP6 complexes under Wnt stimulation and inhibition conditions
Pathway activity assays:
TOP/FOP luciferase reporter assays to measure Wnt pathway activity while modulating TMEM131L levels
Quantification of β-catenin nuclear translocation in relation to TMEM131L expression
Analysis of Wnt target gene expression (e.g., AXIN2, cMYC) in response to TMEM131L manipulation
Degradation kinetics assessment:
Pulse-chase experiments to track LRP6 half-life in the presence/absence of TMEM131L
Lysosomal inhibitor studies to confirm the degradation pathway
Western blot analysis of LRP6 phosphorylation status in relation to TMEM131L levels
Structure-function analysis:
Mutational studies to identify TMEM131L domains critical for Wnt antagonism
Investigation of whether the PapD-L domains implicated in collagen trafficking also participate in Wnt regulation
Deletion constructs to separate TMEM131L's roles in collagen secretion from its Wnt regulatory functions
These approaches can help delineate the mechanistic details of how TMEM131L regulates Wnt signaling and potentially identify novel therapeutic targets for conditions with aberrant Wnt pathway activation.
Researchers working with HRP-conjugated TMEM131L antibodies may encounter several technical challenges. Here are methodological solutions for common issues:
High background signal:
Cause: Insufficient blocking, excessive antibody concentration, or non-specific binding
Solution: Increase blocking time to 2 hours, optimize antibody dilution (try 1:50 initially then adjust), use casein-based blockers instead of BSA, or add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions
Weak or no signal:
Cause: Low TMEM131L expression, epitope masking, or antibody degradation
Solution: Increase sample concentration, optimize antigen retrieval (for IHC), try alternative lysis buffers for membrane protein extraction, verify antibody activity with a positive control, or reduce washing stringency
Non-specific bands in Western blotting:
Cause: Cross-reactivity with related proteins or degradation products
Solution: Increase blocking stringency, add 0.1% SDS to antibody dilution buffer, perform peptide competition assays, or optimize transfer conditions for large proteins (>100 kDa)
Signal variability between replicates:
Cause: Inconsistent technique or sample preparation
Solution: Standardize all procedures, prepare fresh working solutions for each experiment, control incubation temperatures precisely, and use automated washing systems if available
Rapid signal fading in HRP detection:
Cause: Photobleaching or substrate depletion
Solution: Minimize light exposure, use signal enhancers, optimize substrate volume, or switch to more stable HRP substrates
Limited reactivity with certain sample types:
Cause: Species differences or isoform specificity
Solution: Verify TMEM131L sequence homology across species of interest, test antibodies targeting different epitopes, or validate with genetic knockdown controls
The HRP-conjugated TMEM131L antibody is optimized for ELISA applications , so additional optimization may be required when adapting it for other techniques like western blotting or immunoprecipitation.
Systematic evaluation of different TMEM131L antibodies is essential for selecting the optimal reagent for specific research applications. Implement this methodological framework for antibody assessment:
Multi-parameter comparison matrix:
Create a standardized evaluation table comparing key parameters:
| Parameter | Antibody A | Antibody B | Antibody C |
|---|---|---|---|
| Epitope region | N-terminal | PapD-L domain | C-terminal |
| Host species | Rabbit | Mouse | Rabbit |
| Clonality | Polyclonal | Monoclonal | Polyclonal |
| Conjugate | HRP | Unconjugated | FITC |
| Validated applications | ELISA | WB, IF | FACS, IP |
| Species reactivity | Human | Human, Mouse | Human |
| Sensitivity (LOD) | 0.5 ng/mL | 2 ng/mL | 1 ng/mL |
| Specificity (cross-reactivity) | Low | High | Medium |
| Lot-to-lot consistency | Variable | Consistent | Variable |
| Cost-effectiveness | High | Medium | Low |
Side-by-side application testing:
Western blotting: Compare antibodies using identical samples, concentrations, and detection methods; quantify band intensity and background
ELISA: Generate standard curves with recombinant TMEM131L protein; calculate EC50 values and dynamic ranges
Immunofluorescence: Assess staining patterns, signal-to-noise ratios, and co-localization with established markers
Validation in genetic models:
Cross-validation with orthogonal techniques:
Correlate antibody signals with TMEM131L mRNA levels via qPCR
Compare protein detection results with mass spectrometry data
Validate localization patterns with fluorescently tagged TMEM131L
Application-specific optimization matrix:
Document optimal conditions for each antibody across applications:
| Condition | ELISA | Western Blot | IHC |
|---|---|---|---|
| Optimal dilution | 1:20-1:50 | 1:100-1:500 | 1:50-1:200 |
| Blocking agent | 3% BSA | 5% milk | 10% goat serum |
| Incubation time | 2h RT | Overnight 4°C | 3h RT |
| Detection system | TMB substrate | ECL | DAB |
Reproducibility assessment:
Test inter-lot variability by comparing multiple lots of the same antibody
Evaluate reproducibility across different users and laboratories when possible
Assess stability over time with repeated freeze-thaw cycles
This systematic approach enables objective comparison of different TMEM131L antibodies and selection of the optimal reagent for specific research applications.
TMEM131L has been implicated in regulating thymocyte proliferation and shows correlation with immune infiltration in cancer contexts . Researchers can use HRP-conjugated TMEM131L antibodies to explore its immunological functions through these methodological approaches:
Immune cell profiling:
Develop flow cytometry panels (using the same antibody clone in non-HRP format) to assess TMEM131L expression across immune cell populations
Correlate TMEM131L levels with activation states and functional markers
Compare expression in healthy versus disease-associated immune cells
Mechanistic studies in thymocyte development:
Use ELISA with HRP-conjugated antibodies to quantify TMEM131L during stages of thymocyte maturation
Assess how TMEM131L expression changes during positive and negative selection
Correlate TMEM131L levels with key developmental markers and transcription factors
Tumor immune microenvironment analysis:
Functional immune assays:
Investigate how TMEM131L modulation affects:
T cell proliferation and cytokine production
Macrophage polarization and function
Interaction between immune and tumor cells
Targeted pathway investigations:
Explore TMEM131L's relationship with specific immune signaling pathways:
Connection to Wnt signaling in immune contexts
Potential roles in inflammasome activation
Involvement in cytokine receptor trafficking or stability
Therapeutic implications:
Develop strategies to target TMEM131L for immunomodulation
Assess whether TMEM131L status predicts response to immunotherapy
Explore TMEM131L as a biomarker for immune-related adverse events
These research directions could establish TMEM131L as an important regulator of immune function with implications for autoimmunity, cancer immunotherapy, and inflammatory disorders.
TMEM131L has been linked to oxidative stress phenotypes in glioma , suggesting an important functional relationship that warrants further investigation. Researchers can employ these methodological approaches using HRP-conjugated TMEM131L antibodies:
Correlation analysis with oxidative stress biomarkers:
Use ELISA to quantify TMEM131L in relation to:
Lipid peroxidation products (MDA, 4-HNE)
Protein carbonylation levels
GSH/GSSG ratio
SOD and catalase activities
Create multiparameter correlation matrices across patient samples
Functional modulation studies:
Manipulate TMEM131L expression and assess impact on:
Cellular ROS levels using fluorescent probes
Mitochondrial membrane potential
Antioxidant response element (ARE) activation
NRF2 pathway regulation
Targeted transcriptomic analysis:
Following TMEM131L knockdown or overexpression, analyze expression changes in:
Oxidative stress response genes (SOD1/2, CAT, GPX)
ROS-generating enzymes (NOX family)
Mitochondrial genes related to redox balance
Antioxidant pathway components
Co-expression network analysis:
Structure-function studies:
Investigate whether specific domains of TMEM131L are required for its effects on oxidative stress
Determine if the PapD-L domains participate in ROS regulation
Assess post-translational modifications of TMEM131L under oxidative conditions
Therapeutic targeting approaches:
Screen for compounds that modulate TMEM131L-dependent oxidative stress phenotypes
Test antioxidant interventions in models with altered TMEM131L expression
Investigate potential synthetic lethality between TMEM131L status and oxidative stress inducers
The established risk prediction model for oxidative stress-related TMEM131L co-expression genes provides a valuable foundation for these investigations, potentially revealing new therapeutic vulnerabilities in cancers like glioma.
Modern multi-omics approaches can significantly enhance our understanding of TMEM131L function when integrated with antibody-based detection methods. Researchers can implement the following methodological framework:
Integrative proteomics approaches:
Use HRP-conjugated TMEM131L antibodies for immunoprecipitation followed by mass spectrometry to:
Identify novel interaction partners
Map the TMEM131L protein interactome under different cellular conditions
Detect post-translational modifications that regulate function
Compare results with publicly available protein-protein interaction databases
Proteogenomic correlation:
Correlate TMEM131L protein levels (detected by antibody-based assays) with:
mRNA expression from RNA-seq data
Copy number variations affecting the TMEM131L locus
Methylation status of TMEM131L promoter regions
Investigate mechanisms of post-transcriptional regulation
Spatial multi-omics integration:
Combine immunohistochemistry using TMEM131L antibodies with:
Spatial transcriptomics to map expression patterns in tissue contexts
Multiplexed ion beam imaging (MIBI) for simultaneous detection of multiple proteins
Digital spatial profiling for regional quantification of TMEM131L and related pathway components
Single-cell multi-modal analysis:
Integrate single-cell protein detection (using flow cytometry) with:
scRNA-seq to correlate TMEM131L protein and mRNA at single-cell resolution
CITE-seq for simultaneous detection of cell surface markers
Metabolic profiling to connect TMEM131L to cellular metabolic states
Functional genomics integration:
Combine CRISPR screens targeting TMEM131L with antibody-based detection to:
Identify genetic dependencies related to TMEM131L function
Map synthetic lethal interactions
Discover new pathway connections
Clinical multi-omics applications:
Develop prognostic and predictive signatures incorporating:
TMEM131L protein levels measured by standardized assays
Genomic alterations affecting TMEM131L or related pathways
Transcriptomic profiles of TMEM131L-associated genes
Methylation signatures correlating with TMEM131L expression