CD9 mAbs modulate cellular behavior through:
Platelet Activation: Anti-CD9 antibodies (e.g., ALB6, 50H.19) induce GPIIb-IIIa complex association, triggering platelet aggregation .
Cancer Metastasis Inhibition: Clone ALB6 reduces melanoma transendothelial migration by disrupting CD9-VEGFR3 interactions .
Exosome Targeting: CD9 mAbs label exosomes due to tetraspanin enrichment on extracellular vesicles .
Senescent Cell Targeting: CD9mAb-conjugated liposomes enhance rapamycin delivery to CD9-overexpressing senescent fibroblasts .
Biotium’s CD9 antibodies (e.g., 2310.9) are optimized for flow cytometry with bright fluorophores like CF®488A and APC-Astral™813 :
| Conjugate | Excitation/Emission (nm) | Catalog Number |
|---|---|---|
| CF®488A | 490/516 | P015-488A-125 |
| APC-Astral™813 | 633/813 | P015-A813-500 |
R&D Systems’ MAB25292 detects CD9 at 24 kDa in human endothelial cells and platelets . Cusabio’s antibody (CSB-MA004969A1m) shows reactivity at dilutions up to 1:32,000 in WB .
Prognostic Value: High CD9 expression correlates with better outcomes in colorectal cancer (Dako antibody) but poorer survival in ovarian cancer (Abcam EPR2949) .
Therapeutic Potential: KBA1412, a fully human anti-CD9 mAb, demonstrated tumor reduction in preclinical models without thrombotic side effects .
Kling Biotherapeutics’ KBA1412-101 trial (NCT05504213) evaluates this human anti-CD9 mAb in advanced solid tumors :
Mechanisms: Dual action via antibody-dependent cellular cytotoxicity (ADCC) and enhanced immune cell infiltration.
Early mouse mAbs (e.g., ALB6) caused lethal thrombosis in primates, delaying clinical translation for decades . KBA1412 avoids this by targeting a non-platelet CD9 epitope .
This monoclonal CD9 antibody is produced from the hybridoma resulting from the fusion of mouse myeloma cells and splenocytes from mice immunized with recombinant human CD9 antigen protein (amino acids 112-195). The antibody is purified from mouse ascites using protein G and achieves a purity exceeding 95%. This unconjugated CD9 monoclonal antibody aligns with the mouse IgG1 isotype. It is suitable for various applications including ELISA, Western blotting, immunohistochemistry, immunofluorescence, and flow cytometry. Notably, it can only detect the human CD9 protein.
CD9, a tetraspanin, is expressed across all major subsets of leukocytes and is also highly expressed by endothelial cells. Through interactions with other tetraspanins, as well as with diverse transmembrane and intracellular proteins, CD9 plays a crucial role in modulating a range of cellular activities. These include intracellular signaling transduction, proliferation, activation, survival, migration, invasion, adhesion, and diapedesis.
CD9 is a 24 kDa transmembrane glycoprotein belonging to the tetraspanin family, characterized by four hydrophobic transmembrane domains with intracellular N and C termini. The protein plays critical roles in cell-cell adhesion, signal transduction, and membrane protein organization. CD9 is expressed on multiple cell types including platelets, eosinophils, basophils, pre-B cells, activated T cells, and neural cell lines . CD9 monoclonal antibodies are important research tools that allow for specific detection, isolation, and functional modulation of CD9-expressing cells. These antibodies have significantly advanced our understanding of tetraspanin biology, particularly in contexts of cell adhesion, migration, and various pathological conditions where CD9 expression is altered. Notably, CD9 has emerged as a key marker in exosome research, alongside CD63 and CD81, making anti-CD9 antibodies essential for exosome isolation and characterization .
CD9 demonstrates distinctive expression patterns across various cell lineages, making it a valuable marker for cellular identification and functional studies. The protein is prominently expressed on:
Early B cells and pre-B cell lines
Platelets (particularly in alpha-granules)
Eosinophils and basophils
Activated T cells
Neural cell lines
Endothelial cells
CD9 expression is also clinically significant as it appears on approximately 90% of non-T cell acute lymphoblastic leukemia cells and about 50% of chronic lymphocytic and acute myeloblastic leukemia cases . This differential expression pattern makes CD9 antibodies valuable for both research and diagnostic applications in hematological malignancies. Researchers should consider these expression patterns when designing experiments to ensure appropriate positive and negative control samples are included for validation.
For optimal flow cytometry results with CD9 monoclonal antibodies, carefully consider these methodological aspects:
Antibody titration: Determine the optimal concentration through titration experiments. For example, the eBioSN4 antibody can be used at ≤0.5 μg per test (where a test is defined as the amount needed to stain a cell sample in 100 μL final volume) .
Cell preparation: Since CD9 is sensitive to certain enzymatic dissociation methods, use gentle cell dissociation techniques when preparing samples from adherent cultures or tissues to preserve epitope integrity.
Blocking strategy: Implement appropriate blocking (typically with serum matching the secondary antibody species) to minimize non-specific binding, especially when working with heterogeneous populations.
Controls: Always include:
Cell numbers: Empirically determine optimal cell numbers, which typically range from 10^5 to 10^8 cells per test depending on CD9 expression levels in your target population .
When analyzing results, establish gating strategies based on clear positive and negative populations, and consider CD9 expression intensity as potentially biologically significant rather than simply presence/absence.
For successful immunoprecipitation (IP) studies with CD9 monoclonal antibodies, follow these methodological recommendations:
Lysis conditions: Use mild, non-ionic detergents (such as 1% Brij-97 or CHAPS) rather than stronger detergents like SDS or Triton X-100, which can disrupt tetraspanin-enriched microdomains and tetraspanin-partner protein interactions .
Cross-linking considerations: When studying CD9 interactions with other membrane proteins, consider whether to use cross-linking reagents. For example, dithiobis(succinimidyl propionate) has been used effectively to study CD9's interactions with the GPIIb-IIIa complex, revealing that CD9 is not physically associated with other membrane proteins in the resting state .
Sequential immunoprecipitation: For investigating complex formation between CD9 and potential partner proteins, employ sequential immunoprecipitation where the first IP isolates CD9 complexes, followed by disruption of the complexes and a second IP targeting the potential partner protein.
Validation strategy: Confirm specificity by:
These methods have successfully demonstrated important biological findings, such as the physical association between CD9 antigen and the glycoprotein IIb-IIIa complex induced by agonistic anti-CD9 antibodies in platelets .
Various anti-CD9 antibody clones exhibit distinctive functional effects due to differences in their epitope binding and subsequent downstream signaling. This diversity makes clone selection critical for experimental outcomes:
Platelet aggregation effects: Clones such as 50H.19 and ALB6 function as powerful platelet agonists, inducing physical association between CD9 and the glycoprotein IIb-IIIa complex (GPIIb-IIIa). This association occurs independently of thromboxane- and ADP-mediated signaling pathways .
Neutrophil adhesion modulation: Anti-CD9 antibodies can induce rapid increases in neutrophil adhesion to endothelium by acting on the endothelial cell. This effect is not mediated by glycoprotein IIb/IIIa or by leukocyte integrins but likely involves CD9-triggered activation events within endothelial cells .
Epitope competition: Some clones exhibit partial cross-blocking of binding sites, indicating overlapping epitopes. For example, the eBioSN4 monoclonal antibody partially cross-blocks binding of another anti-human CD9 antibody, MM2/57 .
Cell-specific responses: The same antibody clone may induce different responses depending on the cell type, reflecting cell-specific CD9 interaction partners and signaling pathways. For instance, anti-CD9 antibodies induce pre-B cell aggregation through mechanisms distinct from those in platelets .
When designing experiments to probe CD9 function, researchers should select antibody clones based on the specific functional outcome being investigated and validate results with multiple clones to distinguish general CD9 functions from clone-specific effects.
CD9 has emerged as one of the most important markers in exosome research, with anti-CD9 antibodies playing crucial roles in exosome isolation, characterization, and functional studies:
Exosome markers: CD9, CD63, and CD81 are the most commonly used tetraspanin markers for exosome identification. These proteins are enriched in exosomes due to their role in the biogenesis and cargo sorting of these extracellular vesicles .
Multivesicular body targeting: Tetraspanins including CD9 are thought to participate in targeting proteins to multivesicular bodies (MVBs) and subsequently to exosomes. Anti-CD9 antibodies can help track this process in live or fixed cells .
Exosome isolation methods:
Immunoaffinity capture: Anti-CD9 antibodies conjugated to magnetic beads or other solid supports enable specific isolation of CD9-positive exosomes from complex biological fluids
Differential detection: Combining anti-CD9 with antibodies against other exosome markers (CD63, CD81) improves specificity in exosome identification and isolation
Cell-type specificity: While CD9 is broadly expressed on many exosomes, its expression levels vary depending on the cell type of origin. Researchers should be aware that relying solely on CD9 for exosome isolation may bias their samples toward certain exosome subpopulations .
For comprehensive exosome characterization, researchers should combine CD9 detection with other tetraspanin markers and complement antibody-based approaches with biophysical characterization methods (e.g., nanoparticle tracking analysis, electron microscopy).
CD9's involvement in tetraspanin-enriched microdomains and various protein interactions can result in epitope masking, potentially compromising antibody binding and detection. Researchers should implement these strategies to mitigate epitope masking issues:
Multiple antibody clones: Utilize at least two different anti-CD9 antibody clones recognizing distinct epitopes. For instance, when one clone (like eBioSN4) shows reduced binding, an alternative clone targeting a different CD9 epitope might remain accessible .
Sample preparation optimization:
Test various fixation protocols, as overfixation can mask epitopes while insufficient fixation may compromise sample integrity
Evaluate different permeabilization methods for intracellular epitopes, balancing membrane permeability with epitope preservation
Consider mild epitope retrieval methods for formalin-fixed samples
Blocking protocol refinement: Optimize blocking solutions to reduce non-specific binding while ensuring the blocking agent itself doesn't mask CD9 epitopes. Empirically test different blocking agents (BSA, serum, commercial blockers) and concentrations.
Contextual validation: When possible, validate CD9 detection in simplified systems (cell lines, purified exosomes) before application to complex samples like clinical specimens or heterogeneous tissue preparations .
Complementary detection methods: Combine antibody-based detection with non-antibody methods when possible, such as mRNA detection or fusion-protein approaches, to corroborate findings when epitope masking is suspected.
These methodological considerations are essential for accurate CD9 detection, particularly in contexts where CD9 is engaged in multiple molecular interactions that might affect epitope accessibility.
Tetraspanin-enriched microdomains (TEMs) are complex membrane structures requiring rigorous experimental controls when studied using CD9 antibodies:
Detergent selectivity controls:
Cholesterol depletion experiments: Since cholesterol contributes to TEM organization, include methyl-β-cyclodextrin treatments as functional controls to disrupt TEMs and confirm the specificity of observed interactions.
Palmitoylation inhibition: As tetraspanin palmitoylation is critical for TEM formation, include controls with palmitoylation inhibitors or use palmitoylation-deficient CD9 mutants.
Expression level controls:
Establish dose-dependency by analyzing cells with different CD9 expression levels
Use inducible expression systems to control CD9 levels and observe concentration-dependent effects on TEM formation
Secondary antibody controls: For microscopy studies of TEMs, carefully validate secondary antibody specificity and perform controls without primary antibody to exclude non-specific clustering induced by secondary antibodies alone.
Co-localization specificity: When studying CD9 co-localization with partner proteins, include controls with proteins known not to associate with TEMs to establish the specificity of observed co-localization patterns .
Discrepancies in CD9 localization patterns detected by different antibody clones require systematic analysis:
Epitope-specific differences: Different antibody clones may recognize distinct CD9 epitopes that become accessible only in specific conformational states or protein complexes. Document and compare the specific epitopes recognized by each antibody clone based on manufacturer information or epitope mapping studies .
Assay-dependent effects: CD9 detection patterns may vary across methods due to different sample preparation requirements:
Flow cytometry typically uses live or mildly fixed cells
Immunofluorescence requires fixation and potential permeabilization
Western blotting involves denaturation and potential epitope alteration
Validate findings using complementary techniques with the same antibody clone to determine if discrepancies are technique-dependent .
Methodological analysis table:
| Technique | Sample Preparation | Potential Impact on CD9 Detection | Validation Approach |
|---|---|---|---|
| Flow cytometry | Minimal processing | Most native state, but limited spatial resolution | Compare surface vs. permeabilized detection |
| Immunofluorescence | Fixation, permeabilization | Good spatial resolution, but epitope alteration risk | Test multiple fixation protocols |
| Western blot | Denaturation, reduction | Detects total protein, loses conformational epitopes | Compare reducing vs. non-reducing conditions |
| Immunoprecipitation | Detergent lysis | Maintains some protein interactions, disrupts others | Compare mild vs. stringent lysis conditions |
Post-translational modifications: Consider whether discrepancies reflect different CD9 post-translational modifications (glycosylation, palmitoylation) that affect antibody recognition. Include controls such as deglycosylation treatments or palmitoylation inhibitors .
Resolution through combined approaches: When faced with conflicting results, implement a multi-antibody, multi-technique approach, correlating findings with functional outcomes to determine which pattern most accurately reflects biologically relevant CD9 distribution.
Researchers frequently encounter these technical challenges when working with CD9 antibodies:
Sensitivity to fixation conditions: CD9 epitopes can be masked or altered during fixation.
Detergent sensitivity: Inappropriate detergent selection can disrupt tetraspanin-enriched microdomains.
Antibody-induced clustering: Some anti-CD9 antibodies can induce artificial clustering or activation.
Temperature-dependent internalization: CD9 can rapidly internalize upon antibody binding at 37°C.
Solution: Perform binding steps at 4°C when studying surface expression, or include time-course analyses to account for internalization kinetics when working at physiological temperatures.
Clone cross-reactivity: Some anti-CD9 clones may cross-react with other tetraspanins.
Expression level variation: CD9 expression can vary substantially between cell types and under different conditions.
By anticipating these common pitfalls and implementing appropriate technical solutions, researchers can significantly improve the reliability and reproducibility of their CD9 antibody-based studies.
CD9 plays critical roles in various cellular fusion processes, and antibodies against CD9 provide valuable tools for mechanistic investigation:
Gamete fusion studies: CD9 knockout mice show severely reduced female fertility due to impaired sperm-egg fusion. Researchers can use:
Blocking studies with different anti-CD9 antibody clones to determine which epitopes are critical for fusion
Time-lapse imaging with fluorescently-labeled non-blocking anti-CD9 antibodies to track CD9 dynamics during fusion events
Combination approaches with antibodies against fusion partner proteins to elucidate the sequential assembly of fusion complexes
Muscle cell fusion regulation: CD9 and CD81 tightly control muscle cell fusion during regeneration. Methodological approaches include:
Using antibodies that modulate (either enhance or inhibit) fusion to identify functional domains
Immunoprecipitation with anti-CD9 antibodies to isolate fusion-relevant protein complexes at different stages of the fusion process
Correlative light-electron microscopy with immunogold-labeled anti-CD9 antibodies to visualize CD9 localization at fusion sites
Quantitative fusion assays: Develop robust quantification methods combining:
Flow cytometry to measure fusion efficiency in the presence of various anti-CD9 antibodies
Live-cell imaging with fluorescent membrane markers and labeled anti-CD9 antibodies
Biochemical verification of fusion-dependent protein mixing using compartment-specific markers
Therapeutic implications: Anti-CD9 antibodies that modulate fusion events may have applications in fertility treatments, muscle regeneration therapies, and viral infection prevention, as fusion mechanisms share common molecular machinery.
When designing these studies, researchers should carefully select antibody clones based on whether they aim to block CD9 function, track CD9 movement, or isolate CD9-containing complexes without disrupting native interactions.
Investigating the complex dynamics of CD9 interactions with other tetraspanins requires sophisticated methodological approaches:
Advanced microscopy techniques:
Super-resolution imaging: Techniques such as STORM, PALM, or STED can resolve CD9-containing microdomains below the diffraction limit, revealing organization patterns invisible to conventional microscopy
FRET analysis: Using differently labeled antibodies against CD9 and other tetraspanins (CD63, CD81) to measure molecular proximity (<10 nm) in living cells
Single-particle tracking: Following individual CD9 molecules using quantum dot-conjugated Fab fragments to analyze diffusion characteristics within and outside microdomains
Biochemical approaches:
Proteomic analysis of immunoisolated complexes: Using anti-CD9 antibodies for immunoprecipitation followed by mass spectrometry to identify the complete interactome
Blue native PAGE: Preserving native protein complexes to analyze the higher-order assemblies containing CD9
Chemical crosslinking: Using membrane-permeable crosslinkers with subsequent anti-CD9 immunoprecipitation to capture transient interactions
Genetic manipulation strategies:
Domain swapping experiments: Combining antibody detection of chimeric constructs to determine which CD9 domains are critical for microdomain formation
Site-directed mutagenesis: Targeting palmitoylation sites while monitoring antibody accessibility to assess how post-translational modifications affect CD9 incorporation into microdomains
Lipid microdomain analysis:
These methodologies have revealed that tetraspanin-enriched microdomains are distinct from classical lipid rafts and form a specialized type of membrane organization involved in numerous cellular processes including signal transduction, membrane protein trafficking, and cell-cell fusion events.
CD9 antibodies are becoming instrumental in developing exosome-based liquid biopsy approaches for various diseases:
Multiplexed detection platforms: Researchers are developing diagnostic systems combining anti-CD9 antibodies with antibodies against disease-specific exosomal markers. This approach enables:
Microfluidic isolation systems: Novel microfluidic devices utilize anti-CD9 antibodies immobilized on chip surfaces to:
Capture exosomes directly from minimally processed biological fluids
Allow for downstream molecular analysis of captured vesicles
Enable point-of-care testing through miniaturized detection systems
Cancer diagnostics applications: Exosomal CD9 expression profiles may have prognostic value in certain malignancies:
Methodological requirements for clinical translation:
Antibody standardization: Use of consistent, well-validated anti-CD9 clones with established sensitivity and specificity
Sample processing protocols: Development of standardized exosome isolation methods compatible with diverse clinical sample types
Reference standards: Creation of CD9-positive exosome standards for assay calibration and quality control
While promising, researchers must address challenges related to exosome heterogeneity, CD9 expression variability across exosome subpopulations, and standardization of isolation and detection protocols before clinical implementation.
CD9 and other tetraspanins play complex roles in viral infection cycles, and antibodies against CD9 provide valuable tools for mechanistic studies:
Viral entry studies: Several viruses utilize tetraspanin-enriched microdomains for cellular entry. Researchers can:
Use blocking anti-CD9 antibodies to determine whether CD9 is directly involved in viral attachment or entry
Employ non-blocking antibodies for tracking CD9 redistribution during viral entry
Combine antibody approaches with super-resolution microscopy to visualize viral particle co-localization with CD9-enriched domains
Viral assembly and budding: CD9 may participate in viral assembly sites at the plasma membrane. Methodological approaches include:
Immunoprecipitation with anti-CD9 antibodies to identify virus-specific proteins recruited to tetraspanin-enriched domains
Live-cell imaging using fluorescently-labeled anti-CD9 antibodies to track the dynamics of assembly site formation
Electron microscopy with immunogold-labeled antibodies to precisely localize CD9 relative to budding viral particles
Model-specific considerations:
Hepatitis C virus: CD9's association with CD81 (a HCV receptor) makes anti-CD9 antibodies useful for studying receptor complex formation
HIV: CD9 has been implicated in HIV budding, making antibodies valuable for studying late stages of the viral lifecycle
Influenza: Research suggests tetraspanins may organize viral assembly sites, with anti-CD9 antibodies helping to characterize these domains
Therapeutic exploration: Beyond basic research, anti-CD9 antibodies or antibody derivatives may have potential as antiviral agents by:
Disrupting tetraspanin-enriched microdomains required for viral assembly
Blocking specific CD9-viral protein interactions
Triggering CD9 internalization to remove critical viral co-factors from the cell surface
When designing these studies, researchers should carefully characterize how their chosen anti-CD9 antibodies affect normal CD9 functions to distinguish between specific antiviral effects and general cellular perturbations.
When implementing a new CD9 antibody clone in research protocols, systematic validation is essential to ensure reliable and reproducible results:
Specificity validation:
Positive and negative controls: Test the antibody on known CD9-positive cells (platelets, pre-B cell lines) and CD9-negative or knockdown/knockout cells
Western blot analysis: Confirm recognition of a band at the expected molecular weight (~24 kDa) with appropriate controls
Peptide competition: If available, demonstrate signal reduction when pre-incubated with the immunizing peptide
Cross-reactivity assessment: Test against related tetraspanins (CD63, CD81) to confirm specificity
Application-specific validation:
Flow cytometry: Determine optimal concentrations (typically ≤0.5 μg per test) and staining conditions through titration experiments
Immunofluorescence: Optimize fixation and permeabilization protocols to preserve epitope accessibility
Immunoprecipitation: Verify ability to efficiently capture CD9 from lysates prepared with appropriate detergents
Western blotting: Determine optimal conditions (reducing vs. non-reducing, sample preparation)
Functional characterization:
Agonistic/antagonistic activity: Assess whether the antibody induces biological effects (e.g., platelet aggregation, cell adhesion) independent of its detection capability
Epitope mapping: Determine which domain of CD9 is recognized and how this relates to functional outcomes
Cross-blocking studies: Compare with well-characterized CD9 antibody clones to determine epitope relationships
Documentation and reproducibility:
Maintain detailed records of validation experiments
Document lot-to-lot variation when receiving new antibody batches
Consider creating internal reference standards for long-term comparability
These validation steps ensure that experimental outcomes reflect genuine CD9 biology rather than antibody-specific artifacts or technical limitations.
Implementing multiple anti-CD9 antibody clones in integrated research strategies can provide more robust and comprehensive insights:
Complementary epitope targeting:
Use antibodies recognizing different CD9 domains (EC1, EC2, intracellular regions) to build a complete picture of CD9 structure-function relationships
Select clones with non-overlapping epitopes based on cross-blocking studies (e.g., eBioSN4 only partially cross-blocks MM2/57, indicating distinct but potentially overlapping epitopes)
Multi-method validation framework:
Primary discovery: Use one antibody clone for initial observations
Technical validation: Confirm findings with a second clone using the same technique
Methodological validation: Verify results with a different technique using both antibody clones
Functional vs. detection applications:
Separate antibodies used for functional modulation (blocking or activating CD9) from those used for detection to avoid confounding effects
For example, when studying CD9's role in platelet aggregation, use one antibody to induce aggregation and a non-interfering clone targeting a different epitope for detection
Specialized research strategies:
Proximity studies: Use differently labeled anti-CD9 antibody pairs for FRET/BRET assays to study CD9 homodimerization
Trafficking analysis: Combine non-competing antibodies recognizing internal and external epitopes to distinguish surface from internalized CD9 pools
Conformational studies: Employ conformation-sensitive antibodies alongside pan-CD9 antibodies to detect activation-dependent epitope exposure
Documentation and standardization:
Maintain comprehensive records of which clones work optimally for specific applications
Create detailed protocols specifying clone-specific optimization parameters
Consider developing in-house reference standards for inter-experimental comparability