STOM antibodies are immunoreagents designed to detect stomatin, a Band 7/mec-2 family protein involved in transmembrane ion transport regulation . Key features include:
Subcellular localization: Cell membrane, cytoplasmic vesicles, and cytoplasm .
Functional role: Modulation of monovalent cation transport via lipid membranes and interaction with ACCN1/ACCN3 ion channels .
Loss of stomatin localization is linked to hereditary stomatocytosis, a hemolytic anemia .
Antibody validation ensures specificity and reliability. The five pillars of validation, as proposed by international standards, include :
Validation Strategy | Principle | Suitable Applications |
---|---|---|
Genetic (e.g., CRISPR/Cas9) | Target protein elimination/reduction via gene editing; reduced signal = specificity confirmation | WB, IHC, ICC, IP/ChIP |
Orthogonal (e.g., mass spectrometry) | Correlation of antibody signal with antibody-independent quantification methods | WB, IHC, ICC |
Independent Antibody | Comparison of two antibodies targeting non-overlapping epitopes | WB, IHC, ICC, IP/ChIP |
Tagged Protein Expression | Detection of epitope tags expressed at endogenous levels | WB, IHC, ICC |
Immunocapture-MS | Target protein identification via mass spectrometry post-immunocapture | IP/ChIP |
For example, STOM antibody specificity was confirmed in knockout mouse models (Triple KO and Stoml3 KO), where immunostaining vanished in genetically modified tissues .
STOM antibodies have been pivotal in elucidating protein localization and function:
STOM: Enriched in apical dendritic knobs and cilia of OSNs, with vesicular co-localization with Rab11 (an endosomal marker) .
STOML3: Localized to OSN cilia and knobs; absence disrupts STOM ciliary localization .
STOM, also known as stomatin-like protein 2, is an integral membrane protein with 288 amino acid residues that plays essential roles in lipid raft organization and various cellular processes. The significance of STOM lies in its regulatory functions for ion channels and maintenance of cell membrane integrity. It participates in critical cellular processes including cell migration and adhesion .
STOM research has particular relevance for understanding diseases related to membrane defects and ion channel dysregulation. The protein's membrane localization and interaction with the cytoskeleton make it an important target for studies of cellular architecture and signaling pathways. Researchers investigating membrane biology, ion channel regulation, and related pathologies frequently utilize STOM as a model protein for understanding broader membrane protein dynamics .
Researchers have access to several types of STOM antibodies, with polyclonal antibodies being among the most commonly used. Specific examples include:
Polyclonal antibodies: Such as the STOM Rabbit Polyclonal Antibody (CAB6372), which recognizes human, mouse, and rat STOM and is validated for Western blot applications .
Monoclonal antibodies: These offer higher specificity for particular epitopes of the STOM protein.
Species-specific antibodies: Antibodies with reactivity to human, mouse, or rat STOM, with some offering cross-reactivity across multiple species .
The choice between these antibody types depends on the specific research application, with polyclonal antibodies offering broader epitope recognition and monoclonal antibodies providing higher specificity for particular regions of the protein.
Specificity validation for STOM antibodies typically involves multiple complementary approaches:
Immunoblotting with positive control samples (mouse liver, spleen, heart, and rat liver for STOM antibodies)
Comparison of detected molecular weight with the expected size (31.7 kDa for human STOM)
Epitope mapping using recombinant protein fragments
Testing in knockout/knockdown systems to confirm signal reduction
Cross-validation using multiple antibodies targeting different epitopes
For example, the STOM Rabbit Polyclonal Antibody (CAB6372) is validated against recombinant fusion protein containing amino acids 55-288 of human STOM (NP_004090.4), which represents a significant portion of the full protein .
STOM antibodies are validated for several research applications, though the specific validations vary by product. Common applications include:
Western blot (WB): The most common application, typically using dilutions between 1:500 and 1:2000
ELISA: For quantitative detection of STOM in solution
Immunohistochemistry (IHC): For detection in tissue sections
Immunofluorescence (IF): For subcellular localization studies
Immunoprecipitation (IP): For protein-protein interaction studies
Researchers should always verify the validation status for their specific application, as not all antibodies are validated for all possible techniques.
Optimizing Western blot protocols for STOM detection requires attention to several key parameters:
Sample preparation: For membrane proteins like STOM, complete solubilization is crucial. Use buffer systems containing 1-2% SDS or specialized membrane protein extraction buffers.
Running conditions:
Transfer parameters:
For STOM, semi-dry transfer at 15V for 30 minutes or wet transfer at 100V for 1 hour is typically effective
PVDF membranes may provide better results than nitrocellulose for this membrane protein
Antibody conditions:
Detection system:
Enhanced chemiluminescence (ECL) systems work well for STOM detection
For quantitative analysis, consider fluorescence-based detection systems
Proper experimental design with STOM antibodies requires several controls:
Positive tissue controls: Mouse liver, spleen, heart, and rat liver are recommended positive controls for STOM antibody validation .
Negative controls:
Primary antibody omission
Isotype control (non-specific IgG from the same species)
Ideally, STOM-knockout or knockdown samples
Loading controls: For Western blot analysis, include appropriate loading controls:
Pan-membrane protein markers (e.g., Na⁺/K⁺-ATPase) for membrane fraction studies
Standard housekeeping genes (GAPDH, β-actin) for whole-cell lysates
Peptide competition: Pre-incubation of the antibody with excess immunizing peptide should abolish specific signal.
Multiple antibody validation: When possible, confirm findings using a second antibody targeting a different epitope of STOM.
Accurate quantification of STOM expression requires:
Proper normalization strategies:
For Western blots: Normalize to appropriate loading controls
For qPCR analysis of STOM mRNA: Use multiple reference genes (GAPDH, β-actin, 18S rRNA)
Standard curves:
For absolute quantification, generate standard curves using recombinant STOM protein
Ensure standards span the expected range of expression in samples
Image analysis for Western blots:
Use software that corrects for background and performs densitometry
Ensure signal is within the linear range of detection
Analyze multiple independent biological replicates
Complementary approaches:
Validate protein expression findings with mRNA analysis
Consider flow cytometry for cell-by-cell quantification in heterogeneous populations
Method | Advantages | Limitations | Typical Sensitivity |
---|---|---|---|
Western Blot | Semi-quantitative, widely available | Limited throughput | 0.1-1 ng protein |
ELISA | Fully quantitative, high throughput | Requires validated ELISA kit | 10-100 pg/mL |
qPCR (mRNA) | High sensitivity, quantitative | Measures transcript, not protein | Single-copy detection |
Flow Cytometry | Single-cell resolution | Requires cell suspension | Surface proteins |
Non-specific binding is a common challenge with antibodies, including those targeting STOM. Resolution strategies include:
Optimization of blocking conditions:
Test different blocking agents (5% BSA, 5% non-fat milk, commercial blockers)
Increase blocking time (1-2 hours at room temperature or overnight at 4°C)
Antibody dilution adjustment:
Buffer modifications:
Add 0.1-0.3% Triton X-100 or 0.05-0.1% Tween-20 to reduce hydrophobic interactions
Include 5% serum from the secondary antibody species to reduce background
Washing protocol enhancement:
Increase number and duration of washes
Use higher salt concentration (up to 500 mM NaCl) in wash buffers
Pre-adsorption:
For tissues with high endogenous biotin, use avidin/biotin blocking kits
Consider pre-adsorbing antibodies with tissues/cells from species with anticipated cross-reactivity
STOM is primarily localized to the cell membrane with some cytoplasmic presence . For accurate subcellular localization studies:
Sample preparation considerations:
Fixation method significantly impacts membrane protein preservation
For STOM, 4% paraformaldehyde fixation for 10-15 minutes is often optimal
Permeabilization should be gentle (0.1% Triton X-100 for 5-10 minutes)
Co-localization markers:
Include established markers for cell membrane (e.g., Na⁺/K⁺-ATPase)
For lipid raft studies, co-stain with cholera toxin B subunit or caveolin-1
Imaging considerations:
Super-resolution microscopy may be required to resolve lipid raft localization
Z-stack acquisition ensures complete visualization of membrane-associated proteins
Validation approaches:
Confirm localization using subcellular fractionation followed by Western blot
Use epitope-tagged STOM constructs as complementary approach
Contradictory results between antibodies targeting the same protein can stem from several factors:
Epitope differences:
Resolution strategies:
Map the epitopes of each antibody relative to functional domains of STOM
Verify which epitopes might be masked in certain experimental conditions
Consider that post-translational modifications may affect epitope accessibility
Validation approaches:
Use genetic approaches (knockout/knockdown) to confirm specificity
Test antibodies in multiple applications to build confidence
Consider that different results may reflect biological reality (variant forms, modifications)
Reporting recommendations:
Document all antibodies used (catalog numbers, lots, dilutions)
Report all results, even contradictory ones, with potential explanations
Consider publishing validation data alongside main findings
STOM antibodies can be valuable tools for studying protein-protein interactions through several approaches:
Co-immunoprecipitation (Co-IP):
Use STOM antibodies to pull down STOM and associated proteins
Analyze precipitated complexes by mass spectrometry or immunoblotting
Validation requires reversed Co-IP with antibodies against putative binding partners
Proximity ligation assay (PLA):
Combines antibody recognition with DNA amplification to visualize interacting proteins
Requires antibodies raised in different species or directly conjugated to PLA probes
Offers single-molecule sensitivity and spatial resolution
FRET/BRET applications:
For live-cell studies, combine STOM antibody fragments with fluorescent proteins
Enables real-time monitoring of dynamic interactions
Requires careful controls for fluorophore orientation effects
Cross-linking followed by immunoprecipitation:
Chemical cross-linkers stabilize transient interactions before cell lysis
STOM antibodies then isolate STOM-containing complexes
Mass spectrometry identifies cross-linked partners
STOM antibodies are increasingly utilized in disease-related research contexts:
Membrane disorder studies:
STOM antibodies help investigate diseases associated with membrane defects
Quantification of STOM expression serves as a biomarker in certain conditions
Changes in STOM localization may indicate pathological alterations in membrane organization
Ion channel dysfunction research:
STOM's role in regulating ion channels makes it relevant to channelopathies
Antibodies help correlate STOM expression/localization with channel function
Combined electrophysiology and immunolabeling approaches provide functional insights
Cancer biology applications:
Altered STOM expression has been observed in certain cancers
Antibodies enable tissue microarray analysis across tumor types
Correlation of expression with clinical outcomes helps establish prognostic relevance
Neurodegenerative disease research:
Membrane protein dysfunction is implicated in several neurodegenerative conditions
STOM antibodies facilitate investigation of membrane integrity in affected tissues
Post-mortem tissue analysis requires specialized protocols for degradation-sensitive epitopes
Working with STOM across species requires consideration of several factors:
Emerging antibody engineering approaches offer several potential improvements for STOM research:
Single-domain antibodies (nanobodies):
Smaller size allows better access to sterically hindered epitopes
Potential for improved penetration in tissue sections
Greater stability for long-term storage and challenging conditions
Recombinant antibody fragments:
Fab or scFv formats eliminate Fc-mediated artifacts
Production in bacterial systems reduces batch-to-batch variability
Site-specific conjugation enables precise positioning of labels
Bispecific antibodies:
Simultaneous binding to STOM and another protein of interest
Enables novel proximity-based detection strategies
May improve sensitivity for co-localization studies
Engineering for subcellular targeting:
Membrane-permeable antibodies for live-cell applications
Compartment-specific targeting sequences for organelle studies
pH-responsive antibodies for endosomal tracking
Recent developments in antibody validation are enhancing confidence in STOM antibody research:
CRISPR/Cas9 knockout validation:
Generation of STOM-knockout cell lines for definitive specificity testing
Provides gold-standard negative controls for antibody validation
Allows identification of potential cross-reactivity with related proteins
Multi-omic correlation approaches:
Correlation of antibody-based detection with RNA-seq and proteomics data
Triangulation increases confidence in expression patterns
Helps identify discrepancies that may indicate technical issues
Structural validation:
Epitope mapping using hydrogen-deuterium exchange mass spectrometry
Computational prediction of epitope accessibility in different conformational states
Cryo-EM validation of antibody binding to target structure
Community-based validation resources:
Antibody validation databases with user-contributed data
Standardized reporting formats for validation experiments
Pre-competitive collaborations among research institutions
Stomatin is an integral membrane protein found in lipid/protein-rich microdomains of almost all human tissues. It was named after the rare human disease, haemolytic anaemia hereditary stomatocytosis. Stomatin is implicated in signal transduction and cell communication, and it may regulate cation movement through ion channels and transporters .
Stomatin is a 31 kDa monotopic integral membrane protein that is palmitoylated, forms homo-oligomers, and associates with cholesterol-rich membrane domains, also known as lipid rafts . It is part of the SPFH superfamily, which includes stomatin-like proteins, prohibitins, flotillin/reggie proteins, bacterial HflK/C proteins, and erlins . The protein’s structure includes a conserved PHB/SPFH domain, which is essential for its function and association with cholesterol-rich membranes .
The absence of stomatin in red cells is associated with Overhydrated Hereditary Stomatocytosis (OHSt), a condition characterized by the leakage of sodium and potassium ions into and from erythrocytes . However, stomatin knockout mice are viable and do not exhibit stomatocytosis, suggesting that the absence of stomatin in OHSt erythrocytes is due to mistrafficking during terminal erythropoiesis .
The mouse anti-human stomatin antibody is a monoclonal antibody that detects the stomatin protein of human origin. It is used in various applications, including Western blotting, immunoprecipitation, immunofluorescence, immunohistochemistry, and enzyme-linked immunosorbent assay (ELISA) . The antibody is available in both non-conjugated and conjugated forms, including agarose, horseradish peroxidase (HRP), phycoerythrin (PE), fluorescein isothiocyanate (FITC), and multiple Alexa Fluor® conjugates .
Stomatin has been extensively studied for its role in cellular processes and its association with lipid rafts. Recent studies have focused on the structure-function analysis of stomatin, revealing the importance of specific amino acid residues and domains for its function . These studies have shown that stomatin is a cholesterol-binding protein and that its association with cholesterol-rich membranes is crucial for its oligomerization and function .