CEACAM16 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of CEACAM16 Antibody

CEACAM16 antibodies are specialized tools designed to detect and study the carcinoembryonic antigen-related cell adhesion molecule 16 (CEACAM16), a secreted glycoprotein critical for cochlear function. These antibodies are primarily used in auditory system research to investigate CEACAM16’s role in maintaining tectorial membrane (TM) integrity, outer hair cell (OHC) stereocilia attachment, and hearing loss mechanisms . CEACAM16 antibodies are validated for applications such as immunofluorescence, Western blotting, immunohistochemistry (IHC), and enzyme-linked immunosorbent assays (ELISA) .

Applications in Research

CEACAM16 antibodies enable precise detection of the protein in diverse experimental systems:

ApplicationKey Use CasesReferences
ImmunofluorescenceLocalizing CEACAM16 to OHC stereocilia tips and TM in cochlear sections .
Western BlottingDetecting monomeric (~50–75 kDa) and oligomeric (dimer/tetramer) forms .
IHCIdentifying CEACAM16 expression in OHCs vs. inner hair cells (IHCs) .
Flow CytometryQuantifying CEACAM16 surface expression in transfected cells .
ELISAMeasuring extracellular CEACAM16 levels in cell culture media .

Antibody Specificity and Validation

Commercial CEACAM16 antibodies undergo rigorous validation to ensure target specificity:

  • Clone SU-9D5 (Genovac): A mouse monoclonal IgG2b/kappa antibody validated via immunofluorescence and Western blotting in transfected cells. It does not cross-react with other CEACAM family members .

  • Rabbit Monoclonal (R&D Systems): Detects CEACAM16 in HEK293 cells transfected with human CEACAM16 via flow cytometry and IHC, with no signal in non-transfected controls .

  • Polyclonal Antibodies (Merck Millipore): Recognize CEACAM16 in mouse cochlear tissue via IHC and Western blotting, confirmed by blocking assays .

Specificity Testing: Antibodies are screened against unrelated CEACAM proteins (e.g., CEACAM1, CEACAM3, CEACAM6) to exclude cross-reactivity .

Research Findings and Functional Insights

CEACAM16 antibodies have elucidated critical aspects of CEACAM16 biology:

  • Localization: Antibodies show CEACAM16 localizes to OHC stereocilia tips and the TM, where it interacts with α-tectorin to stabilize the TM-OHC linkage .

  • Oligomerization: Western blotting reveals CEACAM16 forms disulfide-linked dimers and tetramers, detected under non-reducing conditions .

  • Hearing Loss Mechanisms: Mutations in CEACAM16 (e.g., T140P) disrupt glycosylation, impairing TM integrity and causing autosomal dominant hearing loss (ADNSHL) .

  • Knockout Models: CEACAM16-null mice exhibit TM structural defects (e.g., absent Hensen’s stripe), leading to spontaneous otoacoustic emissions and cochlear instability .

Challenges and Limitations

  • Oligomer Detection: Monoclonal antibodies struggle to detect higher-order CEACAM16 oligomers in Western blots due to insufficient sensitivity .

  • Species-Specific Reactivity: Most antibodies target human CEACAM16; cross-reactivity with rodent homologs requires separate validation .

  • Experimental Conditions: Oligomer analysis requires reducing agents (e.g., DTT) to disrupt disulfide bonds, complicating native protein studies .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery timelines may vary based on your location and chosen delivery method. Please consult your local distributors for precise delivery estimates.
Synonyms
CEACAM16 antibody; CEAL2Carcinoembryonic antigen-related cell adhesion molecule 16 antibody; Carcinoembryonic antigen-like 2 antibody
Target Names
CEACAM16
Uniprot No.

Target Background

Function
CEACAM16 is essential for normal hearing function. It plays a crucial role in maintaining the structural integrity of the tectorial membrane.
Gene References Into Functions
  1. Research indicates that CEACAM6 promotes cell proliferation through the activation of the cyclin D1/CDK4 pathway. PMID: 26497080
  2. A heterozygous missense mutation, c.505G>A (p.G169R) in exon 3 of the CEACAM16 gene, has been identified in families with autosomal dominant nonsyndromic hearing loss. PMID: 25589040
  3. CEACAM16 is capable of forming complex structures with other tectorial membrane proteins, such as alpha-tectorin and beta-tectorin. These interactions likely influence the physical properties of the tectorial membrane. PMID: 22544735
  4. Studies have identified CEACAM16 as a protein that interacts with alpha-tectorin. It is concentrated at the point where the tectorial membrane attaches to the stereocilia. Mutations in CEACAM16 lead to autosomal dominant nonsyndromic hearing loss at the DFNA4 locus. PMID: 21368133
Database Links

HGNC: 31948

OMIM: 614591

KEGG: hsa:388551

STRING: 9606.ENSP00000385576

UniGene: Hs.456381

Involvement In Disease
Deafness, autosomal dominant, 4B (DFNA4B)
Protein Families
Immunoglobulin superfamily, CEA family
Subcellular Location
Secreted.

Q&A

Basic Questions

  • Q: What is CEACAM16 and what are its key structural features?
    A: CEACAM16 (Carcinoembryonic Antigen-related Cell Adhesion Molecule 16) is a secreted protein belonging to the CEA family. Unlike other CEACAMs, it lacks a transmembrane domain or GPI anchor. The mature human CEACAM16 protein consists of 405 amino acids and contains 2 IgC2-like domains and 2 IgV-like domains. It is one of only five CEACAMs conserved across mice, rats, and humans, with human CEACAM16 sharing approximately 90% amino acid identity with mouse CEACAM16 and 89% with rat CEACAM16 .

  • Q: Where is CEACAM16 primarily expressed and what is its physiological role?
    A: CEACAM16 is specifically expressed in the inner ear and plays a critical role in hearing. Research has identified it as a binding partner for alpha tectorin, a component of the tectorial membrane in the cochlea. CEACAM16 has been localized at the tip of outer hair cell (OHC) bundles in the cochlea, consistent with its role in tectorial membrane function. Notably, specific mutations in CEACAM16 have been linked to autosomal dominant nonsyndromic deafness (ADNSHL) .

Advanced Questions

  • Q: How does CEACAM16 differ from other members of the CEA protein family at the molecular level?
    A: CEACAM16 is unique among the CEA family as it is a secreted molecule lacking a recognizable transmembrane domain or GPI anchor that typically characterizes other family members. While CEACAMs are generally part of the glycosylphosphatidylinositol (GPI)-linked immunoglobulin superfamily, CEACAM16's structure with 2 IgC2-like domains and 2 IgV-like domains gives it distinctive properties. This structural difference explains its secretion into the extracellular space, particularly in the inner ear, where it contributes to the tectorial membrane's structure and function .

  • Q: What molecular interactions has CEACAM16 been demonstrated to engage in, and what techniques have established these interactions?
    A: CEACAM16 has been demonstrated to interact specifically with α-tectorin but not with prestin. This has been established through co-immunoprecipitation experiments using anti-V5 immunoprecipitation in cells cotransfected with V5-tagged Ceacam16 and myc-tagged Tecta (α-tectorin) or GFP-tagged prestin. Western blotting of the immunoprecipitated complexes showed that myc–α-tectorin co-immunoprecipitates with CEACAM16 but prestin does not, confirming a specific interaction between CEACAM16 and α-tectorin. This molecular interaction provides insight into CEACAM16's role in maintaining tectorial membrane integrity and auditory function .

Basic Questions

  • Q: What methods are used to validate the specificity of anti-CEACAM16 antibodies?
    A: Validation of anti-CEACAM16 antibody specificity typically involves multiple complementary techniques. Researchers commonly transfect mammalian cells (such as HEK293T cells) with CEACAM16-expressing plasmids (often with epitope tags like V5 or Flag) and then perform dual immunofluorescent staining with both anti-CEACAM16 and anti-tag antibodies. Colocalization of signals confirms specificity. Additionally, Western blotting of cell lysates from transfected versus non-transfected cells is performed to verify that both anti-tag and anti-CEACAM16 antibodies recognize bands of the same molecular weight. Direct ELISA testing is also used to confirm antibody binding to recombinant CEACAM16 .

  • Q: What are the primary applications for CEACAM16 antibodies in inner ear research?
    A: CEACAM16 antibodies are primarily used in inner ear research for: 1) Immunofluorescence on cochlear samples to localize CEACAM16 protein in relation to stereocilia and the tectorial membrane; 2) Western blotting to detect CEACAM16 expression in tissue lysates; 3) Immunoprecipitation to study protein-protein interactions, particularly with tectorial membrane components like α-tectorin; and 4) Flow cytometry for cell-based expression analysis. These applications help researchers understand CEACAM16's role in normal hearing physiology and investigate the pathological consequences of CEACAM16 mutations .

Advanced Questions

  • Q: How can researchers distinguish between wild-type and mutant CEACAM16 proteins using antibody-based approaches?
    A: Distinguishing between wild-type and mutant CEACAM16 proteins typically requires a multi-method approach. Researchers can use epitope-tagged recombinant constructs (WT versus mutant) transfected into cell lines like HEK293T, followed by comparative analysis using: 1) Quantitative Western blotting to measure expression level differences; 2) ELISA assays to quantify secreted protein amounts in culture medium (as demonstrated for the Arg255Gly mutation, which showed significantly higher levels than WT); 3) Immunofluorescence to assess subcellular localization patterns; and 4) Co-immunoprecipitation to determine if mutations affect binding to interacting partners like α-tectorin. These combined approaches can reveal both functional and expression-level differences between wild-type and mutant CEACAM16 .

  • Q: What are the critical considerations when designing flow cytometry experiments with CEACAM16 antibodies?
    A: When designing flow cytometry experiments with CEACAM16 antibodies, researchers should consider: 1) Appropriate controls including non-transfected cells and irrelevant protein transfections to establish negative control boundaries; 2) Careful antibody titration to determine optimal concentration (typically 3-5 μg/mL for primary antibodies); 3) Selection of compatible fluorophores based on available detection channels (examples include Allophycocyanin-conjugated or NorthernLights 557-conjugated secondary antibodies); 4) Proper staining protocols specific to membrane-associated proteins; and 5) Quadrant marker setting based on control antibody staining. As demonstrated in validation studies, careful attention to these parameters enables accurate detection of CEACAM16-positive populations in transfected cell models .

Basic Questions

  • Q: What are the recommended cell transfection methods for overexpressing CEACAM16 in laboratory studies?
    A: For CEACAM16 overexpression studies, researchers commonly use HEK293T cells transfected with expression vectors containing CEACAM16 cDNA (such as pCMV6-CEACAM16-Flag or pRK5-Flag constructs). Lipofectamine 3000 is a frequently used transfection reagent, though standard transfection protocols (Qiagen) can also be effective. Typically, cells are seeded at approximately 50% confluency one day before transfection and transfected with 6 μg plasmid DNA per 10 cm culture plate. For optimal protein expression, the culture medium is often replaced 6-8 hours post-transfection with DMEM containing antibiotics but no serum. Cells are then harvested 48 hours after transfection for subsequent analysis .

  • Q: What protein extraction methods are recommended when studying CEACAM16 secretion?
    A: Since CEACAM16 is a secreted protein, extraction protocols must address both intracellular and secreted forms. For intracellular protein, researchers typically lyse transfected cells in 2×SDS sample buffer containing protease inhibitors (1mM PMSF and 1× protease inhibitor cocktail). For secreted CEACAM16, the culture medium should be collected 48 hours post-transfection, centrifuged at 4000×rpm for 10 minutes at 4°C, and filtered through 0.22 μm filters. The proteins in the supernatant can be concentrated either by precipitation with trichloroacetic acid (followed by acetone washing) or using ultrafiltration tubes (Millipore). These concentration steps are critical due to the relatively low protein content in culture medium .

Advanced Questions

  • Q: How can researchers effectively quantify differential expression of wild-type versus mutant CEACAM16 proteins?
    A: To quantify differential expression of wild-type versus mutant CEACAM16 proteins, researchers should employ a multi-faceted approach: 1) RT-qPCR to measure relative mRNA expression levels using specific primers (e.g., CEACAM16 forward primer 5′-GACCACCTCAACATCAGCAGCAT-3′, reverse primer 5′-GGTCTTGGTGTTCTTCGCAATACATG-3′) with GAPDH as internal control; 2) Western blot analysis of both cell lysates and concentrated culture medium, with densitometric quantification normalized to housekeeping proteins; 3) ELISA assays specifically designed for human CEACAM16 to quantify secreted protein levels in culture medium; 4) Immunofluorescence with signal intensity quantification. Statistical analysis should include triplicate experiments with appropriate statistical tests (t-tests or ANOVA) to determine significance of observed differences, as exemplified in studies of the Arg255Gly mutation that showed significantly higher expression levels compared to wild-type CEACAM16 .

  • Q: What are the key considerations when designing site-directed mutagenesis experiments to study CEACAM16 variants?
    A: When designing site-directed mutagenesis experiments for CEACAM16 variants, researchers should consider: 1) Selection of an appropriate mutagenesis system, such as QuikChange II Site-Directed Mutagenesis (GE Healthcare); 2) Careful primer design with the mutation centered within complementary primer pairs; 3) Template selection, ideally using a well-characterized wild-type CEACAM16 expression construct (like pRc/CMV-hCEACAM16 or pCMV6-CEACAM16-Flag); 4) Inclusion of epitope tags (Flag, V5) to facilitate detection; 5) Verification of mutations by complete Sanger sequencing of the coding region; 6) Functional validation through comparative expression analysis in transfected cells; and 7) Selection of physiologically relevant mutations based on clinical findings from hearing loss patients. For example, studies have successfully used this approach to generate and characterize the G169R and Arg255Gly variants associated with autosomal dominant nonsyndromic hearing loss .

Basic Questions

  • Q: What are common challenges when performing Western blotting for CEACAM16 and how can they be addressed?
    A: Common challenges in CEACAM16 Western blotting include: 1) Low signal intensity from secreted protein, which can be addressed by using protein concentration methods like ultrafiltration tubes or TCA precipitation; 2) Non-specific bands, which can be minimized by optimizing antibody dilutions (typically 1:1000 for anti-CEACAM16) and blocking conditions (5% non-fat milk for 1-2 hours); 3) Inconsistent loading controls for secreted proteins, which can be managed by loading equal volumes of concentrated media and confirming with Ponceau S staining; 4) Variable expression levels between experiments, which can be controlled by standardizing transfection efficiency and cell density. Additionally, use of epitope-tagged constructs (Flag, V5) can provide alternative detection methods when anti-CEACAM16 antibodies yield suboptimal results .

  • Q: What controls should be included in immunofluorescence experiments involving CEACAM16?
    A: Immunofluorescence experiments for CEACAM16 should include: 1) Non-transfected cells as negative controls to establish background staining levels; 2) Cells transfected with empty vector (e.g., pCMV6) as additional negative controls; 3) Positive controls using cells transfected with epitope-tagged CEACAM16 and stained with both anti-CEACAM16 and anti-tag antibodies to confirm colocalization; 4) DAPI nuclear counterstain for cell identification; 5) Single-stained samples for each fluorophore to set compensation in multi-color experiments; and 6) Secondary antibody-only controls to detect non-specific binding. These controls help distinguish true CEACAM16 staining from background and ensure proper interpretation of subcellular localization patterns, particularly important when comparing wild-type and mutant CEACAM16 proteins .

Advanced Questions

  • Q: How can researchers effectively troubleshoot discrepancies between mRNA expression and protein levels in CEACAM16 studies?
    A: When facing discrepancies between CEACAM16 mRNA and protein levels, researchers should: 1) Verify mRNA quantification by using multiple reference genes beyond GAPDH (e.g., β-actin, 18S rRNA) and checking primer efficiency with standard curves; 2) Assess protein stability by pulse-chase experiments with protein synthesis inhibitors (cycloheximide) to determine if mutations affect protein half-life; 3) Examine post-translational modifications by treating samples with deglycosylation enzymes to determine if glycosylation differences account for expression variations; 4) Investigate subcellular trafficking by performing subcellular fractionation and immunostaining with ER/Golgi markers to identify potential secretory pathway disruptions; 5) Analyze protein aggregation using non-denaturing gels or size-exclusion chromatography. For example, in studies of CEACAM16 mutations, discrepancies were resolved by demonstrating that some mutations affected protein secretion efficiency rather than transcription rates .

  • Q: What strategies can address inconsistent results in protein-protein interaction studies involving CEACAM16?
    A: To address inconsistent results in CEACAM16 protein-protein interaction studies, researchers should implement these strategies: 1) Optimize co-immunoprecipitation conditions by testing different lysis buffers (varying detergent types/concentrations), salt concentrations, and incubation times; 2) Confirm protein expression levels before interaction studies by Western blotting input samples; 3) Use reciprocal immunoprecipitation (e.g., pull down with anti-CEACAM16 and probe for binding partners, then reverse); 4) Employ multiple interaction detection methods such as proximity ligation assays or FRET in addition to co-IP; 5) Include physiologically irrelevant proteins (like prestin) as negative controls; 6) Consider using crosslinking agents for transient interactions; 7) Validate interactions in more physiologically relevant systems when possible. As demonstrated in studies with α-tectorin, careful optimization of these parameters can provide consistent evidence for specific interactions .

Basic Questions

  • Q: What are the characterized mutations in CEACAM16 associated with hearing loss?
    A: Several CEACAM16 mutations have been characterized in connection with autosomal dominant nonsyndromic hearing loss (ADNSHL). The most well-documented mutations include: 1) p.Arg255Gly (c.763A>G), identified in patients with progressive hearing loss affecting primarily the mid-frequency range; 2) p.G169R (c.505G>A), discovered in a Chinese family with ADNSHL; 3) Other missense variants that affect conserved residues in the immunoglobulin-like domains. These mutations are thought to disrupt the normal function of CEACAM16 in maintaining tectorial membrane integrity. Functional studies have shown that these mutations may alter protein expression, secretion levels, or interactions with binding partners like α-tectorin, ultimately compromising the amplification mechanism of the tectorial membrane .

  • Q: How are CEACAM16 knockout or mutation models used in hearing research?
    A: CEACAM16 knockout or mutation models provide valuable tools for understanding the protein's role in hearing pathology. These models allow researchers to: 1) Characterize the effects of CEACAM16 absence or mutations on tectorial membrane morphology using histological and ultrastructural techniques; 2) Measure auditory function through auditory brainstem response (ABR) and distortion product otoacoustic emissions (DPOAE) tests; 3) Analyze age-related progression of hearing loss in longitudinal studies; 4) Evaluate potential therapeutic interventions. In cellular models, researchers transfect HEK293T cells with wild-type or mutant CEACAM16 constructs to study protein expression, secretion, and interactions, providing insights into the molecular mechanisms of hearing loss caused by CEACAM16 mutations .

Advanced Questions

  • Q: What experimental approaches can differentiate between pathogenic and benign variants of CEACAM16?
    A: Differentiating between pathogenic and benign CEACAM16 variants requires a comprehensive experimental approach: 1) Functional secretion assays comparing wild-type and variant proteins in transfected cell models, with quantification by ELISA and Western blotting of culture medium (pathogenic variants often show altered secretion patterns); 2) Protein-protein interaction studies using co-immunoprecipitation to assess binding with known partners like α-tectorin; 3) Structural predictions and molecular modeling based on the immunoglobulin-like domains to predict impact on protein folding; 4) Species conservation analysis across evolutionarily distant organisms to identify critical residues; 5) Cell-based localization studies using immunofluorescence to detect subcellular distribution abnormalities; 6) Co-segregation analysis in families with hearing loss; and 7) Population frequency data to establish rarity of variants. For example, the Arg255Gly variant was determined to be pathogenic based on its altered secretion profile (higher extracellular levels) despite normal subcellular localization .

  • Q: How can researchers design experiments to investigate the temporal and spatial expression patterns of CEACAM16 during inner ear development?
    A: To investigate CEACAM16's temporal and spatial expression during inner ear development, researchers should employ: 1) Time-course RNA extraction from cochlear tissues at different developmental stages, followed by RT-qPCR with primers specific to CEACAM16 (forward: 5′-GACCACCTCAACATCAGCAGCAT-3′, reverse: 5′-GGTCTTGGTGTTCTTCGCAATACATG-3′); 2) In situ hybridization on cochlear sections from different developmental timepoints to visualize mRNA expression patterns; 3) Immunohistochemistry and immunofluorescence on cochlear whole-mounts and sections using validated anti-CEACAM16 antibodies, with Z-stack confocal microscopy to create 3D images of protein localization; 4) Co-labeling with markers for specific cochlear structures (actin for stereocilia, α-tectorin for tectorial membrane); 5) Western blotting of tissue lysates from different developmental stages to quantify protein expression; 6) Single-cell RNA sequencing to identify cell-specific expression patterns. This comprehensive approach would provide insights into CEACAM16's role during critical periods of inner ear development and maturation .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.