IGSF3 belongs to the EWI subfamily of immunoglobulin-like proteins, containing eight C2-type Ig-like domains . It interacts with tetraspanins (e.g., CD9) and integrins, modulating cell migration, apoptosis, and barrier function . Deficiencies in IGSF3 have been linked to chronic obstructive pulmonary disease (COPD) and hepatocellular carcinoma (HCC) .
The HRP-conjugated antibody is a rabbit polyclonal antibody raised against specific regions of human IGSF3 (e.g., AA 111–399) . HRP conjugation allows colorimetric detection via chromogenic substrates (e.g., TMB), enhancing sensitivity in ELISA and related assays .
The HRP-conjugated IGSF3 antibody is optimized for ELISA, enabling precise quantification of IGSF3 levels in serum, cell lysates, or tissue homogenates . Its specificity for the AA 111–399 region ensures minimal cross-reactivity with other proteins .
COPD: IGSF3 deficiency disrupts sphingolipid metabolism and barrier function in lung cells, exacerbating injury repair . The antibody aids in studying these mechanisms.
HCC: IGSF3 overexpression correlates with NF-κB activation, promoting tumor migration and invasion. HRP-conjugated antibodies facilitate biomarker validation in HCC models .
A chromosomal translocation disrupting IGSF3 led to severe emphysema, highlighting its role in lung cell adhesion and repair . IGSF3 interacts with CD9 and integrin β1, maintaining membrane integrity .
IGSF3 is upregulated in HCC tissues and cell lines. Silencing IGSF3 inhibits NF-κB signaling, reducing tumor growth in vitro and in vivo . The HRP-conjugated antibody supports its validation as a diagnostic/prognostic marker.
Supplier | Catalog No. | Immunogen Region | Host | Applications | Dilution |
---|---|---|---|---|---|
Antibodies-Online | ABIN7156109 | AA 111–399 | Rabbit | ELISA | N/A |
Abbexa Ltd | N/A | AA 111–399 | Rabbit | ELISA | Optimal dilution determined empirically |
Note: Both antibodies target the same epitope but differ in supplier-specific optimization .
Therapeutic Targeting: Inhibiting IGSF3-NF-κB interactions may offer strategies for HCC treatment .
Cross-Species Studies: Mouse IGSF3 shares 92% homology with human IGSF3, enabling preclinical model validation .
Method Optimization: HRP-conjugated antibodies require careful dilution to avoid background noise in ELISA .
Assay Genie (CAB12449): Detailed antibody specifications for IGSF3 detection.
Antibodies-Online (ABIN7156109): HRP-conjugated rabbit polyclonal antibody for ELISA.
PMC (PMC7453886): IGSF3’s role in COPD and sphingolipid metabolism.
PMC (PMC7186720): IGSF3 as an HCC biomarker and NF-κB pathway activation.
Abbexa Ltd: Technical details for HRP-conjugated IGSF3 antibody.
IGSF3 (Immunoglobulin Superfamily Member 3) is a membrane-localized protein with a canonical form of 1194 amino acid residues and a molecular mass of approximately 135.2 kDa in humans. It belongs to the immunoglobulin superfamily and is also known by several synonyms including LCDD, V8, glu-Trp-Ile EWI motif-containing protein 3, and EWI-3 . IGSF3 has drawn research interest due to its high expression in placenta, kidney, and lung tissues, and its association with lacrimal duct defects . The protein undergoes glycosylation as a post-translational modification, and up to two different isoforms have been reported . Given its membrane localization and tissue-specific expression patterns, IGSF3 is studied for potential roles in cellular adhesion, signaling, and tissue development. Researchers target this protein to understand its biological functions and potential implications in developmental disorders and disease processes.
HRP-conjugated IGSF3 antibodies are primarily optimized for ELISA (Enzyme-Linked Immunosorbent Assay) applications . The horseradish peroxidase conjugation provides a direct enzymatic detection method that eliminates the need for secondary antibodies, simplifying workflows and potentially reducing background signal. While ELISA is the main application, researchers should be aware that unconjugated IGSF3 antibodies have been validated for additional techniques including Western Blot (WB), Flow Cytometry (FCM), Immunohistochemistry (IHC), and Immunofluorescence (IF) . When considering using HRP-conjugated antibodies for applications beyond ELISA, researchers should conduct preliminary validation studies to confirm suitability, as the conjugation could potentially affect binding characteristics in some contexts.
While the canonical molecular weight of IGSF3 is reported as 135.2 kDa based on amino acid sequence , Western blot detection typically reveals a band at approximately 200 kDa . This significant difference between theoretical and observed molecular weight is attributed to post-translational modifications, particularly glycosylation, which is known to occur in IGSF3 . When performing Western blot analysis, researchers should expect to observe IGSF3 at approximately 200 kDa under non-reducing conditions . It's important to note that some studies specifically indicate detection under non-reducing conditions only , suggesting that the protein's conformation and epitope accessibility may be significantly altered under reducing conditions, potentially affecting antibody recognition.
Based on expression data and validated research applications, human lung tissue and the A549 human lung carcinoma cell line have been demonstrated as reliable biological sources for IGSF3 detection . IGSF3 shows high expression in placenta, kidney, and lung tissues , making these primary tissues valuable for studying endogenous expression. For cell culture models, A549 cells have been validated for IGSF3 expression using both flow cytometry and Western blot techniques . When selecting experimental models, researchers should consider these validated sources while recognizing that IGSF3 expression may vary with cell state, culture conditions, and experimental manipulations. For novel tissue or cell line applications, preliminary validation of IGSF3 expression levels is recommended before proceeding with extensive experimental work.
Sample preparation for IGSF3 detection requires careful consideration of the protein's membrane localization and post-translational modifications. For Western blot applications, non-reducing conditions appear critical for optimal detection . Researchers should use lysis buffers containing appropriate detergents (such as NP-40 or Triton X-100) to efficiently solubilize membrane proteins while preserving epitope integrity. When preparing samples, maintain protein samples at 4°C and include protease inhibitors to prevent degradation. For ELISA applications with HRP-conjugated antibodies, sample dilution optimization is essential, as both over-dilution (leading to weak signals) and under-dilution (causing high background) can compromise results.
The following sample preparation protocol is recommended for Western blot applications:
Harvest cells/tissue and wash in cold PBS
Lyse in membrane protein extraction buffer (150 mM NaCl, 50 mM Tris-HCl pH 7.4, 1% NP-40, protease inhibitor cocktail)
Incubate on ice for 30 minutes with occasional vortexing
Centrifuge at 14,000g for 15 minutes at 4°C
Collect supernatant and determine protein concentration
Mix with non-reducing sample buffer (no DTT or β-mercaptoethanol)
Do not heat samples before loading onto gels
For ELISA applications with HRP-conjugated antibodies, maintain samples in buffers compatible with the assay format and avoid detergents that might interfere with antibody-antigen binding.
Optimal antibody dilution is application-specific and should be determined experimentally for each lot of antibody. For HRP-conjugated IGSF3 antibodies in ELISA applications, a recommended starting dilution range is 1:1000 to 1:5000 . Performing a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000, 1:10000) during initial optimization can help identify the dilution that maximizes specific signal while minimizing background.
For reference, unconjugated IGSF3 antibodies have been used at the following concentrations in other applications:
When using HRP-conjugated antibodies in applications beyond their primary recommended use, researchers should start with more conservative dilutions (higher antibody concentration) and optimize based on signal-to-noise ratios. Remember that over-dilution can lead to false negatives, while insufficient dilution can increase non-specific binding and background.
Robust experimental design requires appropriate controls to validate results and ensure reliable interpretations. When using HRP-conjugated IGSF3 antibodies, the following controls should be considered:
Positive control: Include samples known to express IGSF3, such as human lung tissue or A549 cells
Negative control: Include samples known not to express IGSF3, or samples where IGSF3 expression has been knocked down
Isotype control: For flow cytometry applications, include an appropriate isotype control antibody with HRP conjugation to determine background levels
Blocking peptide control: Where available, use a specific blocking peptide corresponding to the immunogen to confirm antibody specificity
Secondary antibody-only control: For troubleshooting, include a control lacking primary antibody to assess background from the detection system
Loading control: For Western blot applications, include detection of a housekeeping protein to normalize for loading variations
Recombinant protein standard: Where possible, include purified IGSF3 protein as a positive control and for quantification purposes
These controls help distinguish specific signals from background and validate the performance of HRP-conjugated IGSF3 antibodies in experimental workflows.
ELISA protocols using HRP-conjugated IGSF3 antibodies can be optimized through careful attention to several key parameters:
Coating optimization: For sandwich ELISA, use a validated capture antibody (typically unconjugated) at 1-5 μg/mL in coating buffer (usually carbonate-bicarbonate pH 9.6). For direct ELISA, coat plates with purified antigen or sample at optimized concentration.
Blocking optimization: Test different blocking agents (BSA, milk proteins, commercial blockers) at 1-5% concentration to identify the formulation that minimizes background without interfering with specific binding.
Sample preparation: Dilute samples in appropriate buffers that maintain protein stability while minimizing matrix effects.
Antibody incubation: Optimize both concentration (as described in section 2.2) and incubation time (typically 1-2 hours at room temperature or overnight at 4°C).
Washing optimization: Determine optimal washing buffer composition (typically PBS-T) and number of wash cycles to remove unbound antibody while preserving specific interactions.
Substrate selection: Choose an appropriate HRP substrate based on required sensitivity (TMB, ABTS, or chemiluminescent substrates).
Signal development and kinetics: Monitor the development of signal over time to determine optimal substrate incubation period before stopping the reaction.
The following table summarizes key optimization parameters for ELISA using HRP-conjugated IGSF3 antibodies:
Parameter | Recommendation | Optimization Range |
---|---|---|
Capture antibody | 2 μg/mL in carbonate buffer pH 9.6 | 1-5 μg/mL |
Blocking agent | 3% BSA in PBS | 1-5% BSA, milk alternatives |
Sample dilution | Application-dependent | Serial dilutions recommended |
HRP-conjugated IGSF3 antibody | 1:2000 in blocking buffer | 1:1000-1:5000 |
Incubation time | 2 hours at RT or overnight at 4°C | 1-3 hours RT, 4-16 hours at 4°C |
Wash cycles | 4 × 300 μL PBST (0.05% Tween-20) | 3-5 wash cycles |
Substrate | TMB | TMB, ABTS, OPD, or chemiluminescent |
Development time | 15 minutes in dark | 5-30 minutes |
IGSF3 has been reported to have up to two different isoforms , which can significantly impact antibody selection and experimental outcomes. When designing experiments, researchers should consider which isoform(s) are relevant to their research question and select antibodies accordingly. The HRP-conjugated IGSF3 antibody (ABIN7156109) targets amino acids 111-399 , which may have different accessibility or presence in various isoforms.
Key considerations for addressing isoform variability include:
Epitope mapping: Review the specific epitope targeted by the antibody and determine whether this region is present in all isoforms of interest.
Isoform-specific detection: For experiments requiring discrimination between isoforms, select antibodies that target unique regions or supplement with molecular techniques like RT-PCR with isoform-specific primers.
Molecular weight verification: Different isoforms may exhibit distinct molecular weights on Western blots; document and characterize these differences in your experimental system.
Tissue/cell specificity: Determine whether different isoforms show tissue or cell-specific expression patterns that might affect your experimental design.
Functional significance: Consider whether different isoforms may have distinct functional roles that could impact biological interpretations of your results.
When using HRP-conjugated IGSF3 antibodies, preliminary experiments to characterize which isoforms are detected in your specific experimental system will strengthen the validity and interpretation of results.
The HRP-conjugated IGSF3 antibody (ABIN7156109) is a polyclonal antibody raised in rabbits . Understanding the differences between polyclonal and monoclonal antibodies is crucial for experimental design and interpretation:
Polyclonal IGSF3 Antibodies:
Recognize multiple epitopes on the IGSF3 protein, potentially increasing detection sensitivity
May provide more robust detection across different experimental conditions and applications
Can be less affected by minor changes in protein conformation or post-translational modifications
May exhibit higher batch-to-batch variability, requiring validation across lots
Generally exhibit higher background due to potential cross-reactivity
Monoclonal IGSF3 Antibodies:
Target a single epitope with high specificity
Provide more consistent results with lower batch-to-batch variability
May be more sensitive to changes in protein conformation or post-translational modifications that affect the specific epitope
Often have lower background signal in applications like IHC and IF
May have reduced sensitivity compared to polyclonals in some applications
For applications using HRP-conjugated IGSF3 polyclonal antibodies, researchers should:
Validate specificity through appropriate controls
Consider epitope availability in different sample preparation conditions
Be aware that detection may represent multiple epitopes on the protein
Validate new antibody lots before use in critical experiments
The choice between polyclonal and monoclonal antibodies should align with experimental goals, balancing the need for sensitivity, specificity, and reproducibility.
Multiplexed detection allows simultaneous analysis of IGSF3 and other proteins of interest, providing insights into co-expression, co-localization, or functional relationships. When incorporating HRP-conjugated IGSF3 antibodies into multiplexed approaches, consider the following strategies:
For Western Blot Multiplexing:
Sequential detection: Strip and reprobe membranes, being mindful that stripping can reduce signal intensity for subsequent detections
Dual-color chemiluminescence: Use HRP-conjugated IGSF3 antibody with another antibody conjugated to a different enzyme (e.g., alkaline phosphatase) and differential substrates
Fluorescence multiplexing: Consider fluorescently labeled alternatives to HRP-conjugated antibodies for true multiplexing capabilities
For Flow Cytometry:
Use HRP-conjugated IGSF3 antibody alongside fluorescently-labeled antibodies against other targets
Consider fluorescent versions of IGSF3 antibodies (e.g., Alexa Fluor® 488-conjugated) for better multiplexing compatibility
For Immunohistochemistry/Immunofluorescence:
For brightfield IHC, use HRP-conjugated IGSF3 antibody on sequential sections rather than true multiplexing
For IF, fluorescent alternatives to HRP-conjugation provide better multiplexing options
When designing multiplexed experiments, careful consideration of antibody compatibility, signal separation, and appropriate controls is essential. Cross-reactivity between antibodies, spectral overlap, and signal intensity balancing are common challenges that require optimization for successful multiplexed detection.
Thorough validation of antibody specificity is essential for reliable research results. For HRP-conjugated IGSF3 antibodies, consider these validation approaches:
Western blot characterization: Confirm detection at the expected molecular weight (~200 kDa) and verify band pattern consistency across multiple sample types
Positive and negative controls: Test antibody performance in tissues/cells with known high expression (lung, placenta, kidney) versus those with low/no expression
Peptide competition assay: Pre-incubate antibody with excess immunizing peptide (recombinant IGSF3 amino acids 111-399) to demonstrate signal reduction
Genetic knockdown/knockout: Validate antibody specificity using IGSF3 siRNA knockdown or CRISPR knockout models to confirm signal reduction
Cross-species reactivity: Assess performance across species to confirm specificity aligns with expected evolutionary conservation
Orthogonal detection methods: Compare results with alternative detection methods (e.g., mass spectrometry) or independent antibodies targeting different IGSF3 epitopes
Application-specific validation: Validate specifically for each intended application (ELISA, WB, FC), as performance may vary across applications
Documentation of these validation steps strengthens the reliability of research findings and should be included in publications to demonstrate antibody performance rigor.
IGSF3 Western blot analysis may reveal unexpected band patterns for several reasons, requiring careful interpretation:
Higher molecular weight than predicted: IGSF3 typically appears at ~200 kDa despite a predicted size of 135.2 kDa , primarily due to extensive glycosylation . This size discrepancy is normal and expected.
Multiple bands: May indicate:
Unexpected band sizes: Could result from:
Alternative splicing variants
Protein degradation during sample preparation
Sample preparation conditions affecting protein migration
Cross-reactivity with related proteins
To properly interpret unexpected band patterns:
Compare to literature: Confirm if observed patterns match published IGSF3 Western blot results
Reduce protein degradation: Use fresh samples, work at 4°C, and include protease inhibitors
Optimize sample preparation: Pay special attention to non-reducing conditions, which appear critical for optimal IGSF3 detection
Validate with controls: Include positive control samples (human lung tissue) for comparison
Glycosidase treatment: Consider enzymatic deglycosylation to confirm glycosylation contribution to observed molecular weight
Epitope mapping: Determine if the antibody recognizes regions present in all potential isoforms or splice variants
Remember that IGSF3 is a membrane protein with extensive post-translational modifications, which can significantly impact its behavior in Western blot applications.
When encountering weak or inconsistent signals with HRP-conjugated IGSF3 antibodies, systematic troubleshooting can identify and resolve underlying issues:
Antibody concentration optimization:
Sample preparation refinement:
Detection system enhancement:
Use high-sensitivity HRP substrates (enhanced chemiluminescence)
Extend exposure times for Western blot or substrate incubation for ELISA
Consider signal amplification systems (biotin-streptavidin)
Ensure HRP activity has not been compromised during storage
Experimental conditions optimization:
Reduce washing stringency if signal is weak
Optimize blocking conditions to prevent over-blocking
Adjust incubation temperature (room temperature vs. 4°C)
Verify buffer compatibility with HRP activity
Antibody storage and handling:
Avoid repeated freeze-thaw cycles
Store according to manufacturer recommendations
Check for precipitation or contamination
Verify antibody hasn't exceeded recommended shelf life
If signal remains problematic after these optimizations, consider alternative antibody formats or detection methods, such as unconjugated primary antibodies with separate HRP-conjugated secondary antibodies for potential signal amplification.
Cross-reactivity and high background signal can compromise experimental results when working with HRP-conjugated IGSF3 antibodies. These issues can be addressed through several strategies:
Optimize blocking conditions:
Test different blocking agents (BSA, casein, commercial blockers)
Increase blocking time or concentration if background is high
Use the same blocking agent in antibody dilution buffer
Antibody dilution optimization:
Increase antibody dilution if background is high
Prepare antibody dilutions in fresh blocking buffer
Pre-absorb antibody with relevant tissues/cell lysates to reduce non-specific binding
Washing optimization:
Increase number of wash steps (5-6 washes instead of 3)
Extend wash durations (5-10 minutes per wash)
Adjust detergent concentration in wash buffers (0.05-0.1% Tween-20)
Reduce non-specific interactions:
Add 0.1-0.5% non-ionic detergent to antibody dilution buffer
Include 5% normal serum from the same species as samples
Consider adding 0.1-1% BSA to antibody dilution buffer
Validate specificity:
Test antibody on samples known to be negative for IGSF3
Perform peptide competition assays to confirm specific binding
Compare patterns across different applications and sample types
Control for endogenous peroxidase activity (for tissue samples):
Pre-treat samples with H₂O₂ to quench endogenous peroxidase
Use specific peroxidase blocking reagents before antibody incubation
Systematic optimization of these parameters can significantly improve signal-to-noise ratio and reduce cross-reactivity when working with HRP-conjugated IGSF3 antibodies.
While the HRP-conjugated IGSF3 antibody is primarily recommended for ELISA applications , researchers interested in flow cytometry should consider the following critical factors if exploring this application or when using alternative IGSF3 antibody formats:
Cell preparation optimization:
Ensure single-cell suspensions with high viability
Optimize fixation protocols to maintain epitope accessibility
Consider membrane permeabilization requirements based on epitope location
Antibody format selection:
Controls for flow cytometry:
Staining protocol optimization:
Titrate antibody concentration to determine optimal signal-to-noise ratio
Optimize incubation time and temperature
Include washing steps to reduce background
Instrument setup and analysis:
Set appropriate voltage and compensation
Use proper gating strategies to identify IGSF3-positive populations
Consider co-staining with markers that define relevant cell subpopulations
Fluorescence values from flow cytometry experiments with A549 cells have demonstrated that IGSF3 is detectable on the cell surface , providing a useful positive control for validation studies. When establishing new flow cytometry protocols for IGSF3 detection, incremental optimization of each parameter will contribute to robust and reproducible results.
IGSF3, as a member of the immunoglobulin superfamily localized to the cell membrane , can be studied in the context of protein complexes and interactions using several approaches:
Co-immunoprecipitation (Co-IP):
Use IGSF3 antibodies to pull down IGSF3 and associated proteins
Western blot analysis can then identify interaction partners
Consider crosslinking approaches to stabilize transient interactions
Optimize lysis conditions to maintain membrane protein complexes
Proximity labeling approaches:
Combine IGSF3 antibodies with proximity labeling techniques (BioID, APEX)
This can identify proteins in close proximity to IGSF3 in living cells
Immunofluorescence co-localization:
FRET/BRET analysis:
Combine fluorescently-labeled IGSF3 antibodies with antibodies against potential partners
Measure energy transfer to assess close molecular proximity
Membrane protein complex isolation:
Use detergent-resistant membrane isolation techniques
Antibody-based affinity purification of intact complexes
Blue native PAGE for analysis of intact complexes
Proteomic approaches:
Antibody-based purification followed by mass spectrometry
Cross-reference findings with predicted interaction networks
These approaches can provide insights into IGSF3's role in membrane organization, signaling complexes, and cellular communication pathways, particularly in tissues with high expression such as lung, placenta, and kidney .
IGSF3 research has potential implications in several disease contexts, particularly given its association with lacrimal duct defects . Researchers using IGSF3 antibodies in disease-related studies should consider:
Developmental disorders:
Investigation of IGSF3 expression in models of lacrimal duct development
Correlation of IGSF3 expression patterns with developmental abnormalities
Potential genetic screening for IGSF3 variants in relevant patient populations
Cancer research:
Immunological studies:
Examination of IGSF3's potential roles in immune cell interactions
Investigation of expression changes during inflammatory responses
Potential involvement in immune cell recruitment or activation
Tissue-specific pathologies:
Biomarker development:
Evaluation of IGSF3 as a potential biomarker in relevant disease contexts
Development of quantitative assays using HRP-conjugated antibodies
Correlation with disease progression or treatment response
Research in these emerging areas requires careful validation of antibody specificity in the particular disease model or tissue context. Researchers should consider combining antibody-based detection with genetic approaches (e.g., RNA-seq, genetic manipulation) to build comprehensive understanding of IGSF3's role in disease processes.
Researchers working with IGSF3 antibodies, particularly HRP-conjugated versions, should adopt these best practices to ensure robust and reproducible results:
Comprehensive validation: Validate antibody specificity through multiple approaches including Western blot, positive/negative controls, and when possible, genetic manipulation of IGSF3 expression.
Application-specific optimization: Optimize protocols specifically for each experimental application, recognizing that conditions optimal for ELISA may differ from those for Western blot or other techniques.
Appropriate controls: Include all necessary experimental controls, including isotype controls, blocking peptide controls, and positive/negative sample controls.
Documentation and reporting: Maintain detailed records of antibody specifications (catalog number, lot, concentration) and experimental conditions for reproducibility and transparent reporting in publications.
Sample preparation considerations: Pay special attention to membrane protein extraction methods and non-reducing conditions for Western blot applications .
Isoform awareness: Consider the impact of IGSF3 isoforms and post-translational modifications on experimental results and interpretations.
Cross-validation: When possible, confirm key findings using alternative antibodies targeting different IGSF3 epitopes or orthogonal detection methods.
Storage and handling: Follow manufacturer recommendations for antibody storage, handling, and working dilution preparation to maintain antibody performance over time.
By implementing these best practices, researchers can maximize the scientific value of experiments using IGSF3 antibodies while ensuring reliability and reproducibility of their findings.
Effective troubleshooting requires a systematic approach to identify and address factors affecting experimental outcomes. For HRP-conjugated IGSF3 antibodies, consider this structured optimization process:
Establish baseline performance:
Systematic parameter optimization:
Modify one variable at a time (antibody concentration, incubation time, etc.)
Document the effect of each change on signal and background
Build an optimization matrix to identify optimal conditions
Sample-specific adjustments:
Adapt protocols for specific sample types (cell lines vs. tissues)
Optimize lysis and extraction methods for membrane proteins
Consider sample-specific interfering factors
Signal development optimization:
Test different HRP substrates for optimal sensitivity
Optimize substrate incubation time and concentration
Determine linear range of detection for quantitative applications
Application-specific considerations:
Documentation and standardization:
Create detailed protocols capturing all optimized parameters
Standardize positive controls across experiments
Establish acceptance criteria for experimental validity