CD14 Antibody, Biotin is widely used in:
In a baboon model of Escherichia coli sepsis, anti-CD14 treatment:
Improved survival: 2/5 animals survived to 7 days vs. 0/5 controls .
Reduced coagulation activation:
Enhanced fibrinolysis:
Cytokine suppression: Decreased TNF-α, IL-6, and IL-1β by >50% in survivors .
Complement inhibition: Reduced C3b, C5a, and sC5b-9 levels (P < 0.05) .
CD14 is a 53-55 kDa GPI-linked glycoprotein that functions primarily as a pattern recognition receptor. It is expressed strongly on the surface of monocytes, weakly on granulocytes, and on most tissue macrophages and some dendritic cells. The canonical human CD14 protein consists of 375 amino acid residues with a molecular weight of approximately 40.1 kDa before post-translational modifications. CD14 exists in both membrane-bound and soluble forms, with soluble CD14 (sCD14) found in serum and in the urine of some nephrotic patients. It is notably expressed in various tissues including the colon and endometrium, where it plays important roles in immune surveillance and response to microbial components .
CD14 serves as a critical coreceptor for bacterial lipopolysaccharide (LPS), forming a complex with LPS-binding protein (LBP) to deliver monomeric LPS to the LY96/TLR4 complex. This interaction mediates the innate immune response to bacterial LPS through signaling pathways involving MyD88, TIRAP, and TRAF6, ultimately leading to NF-kappa-B activation, cytokine secretion, and inflammatory responses. Beyond its role in LPS recognition, CD14 also functions as a coreceptor for TLR2:TLR6 heterodimers in response to diacylated lipopeptides and for TLR2:TLR1 heterodimers in response to triacylated lipopeptides. Additionally, CD14 binds electronegative LDL (LDL(-)) and mediates cytokine release induced by LDL(-), suggesting roles in atherosclerosis and other inflammatory conditions .
Biotin-conjugated CD14 antibodies offer significant advantages in research due to the exceptionally high affinity between biotin and streptavidin/avidin (Kd ≈ 10^-15 M), which is one of the strongest non-covalent interactions in biology. This property enables powerful signal amplification through secondary detection systems, enhancing sensitivity in assays such as flow cytometry, immunohistochemistry, and ELISA. The biotin-streptavidin system provides remarkable flexibility in experimental design, allowing researchers to use various streptavidin-conjugated detection reagents (fluorophores, enzymes, or magnetic particles) with the same biotinylated primary antibody. Importantly, when properly performed, the biotinylation process preserves the antibody's specificity and binding characteristics while adding the biotin tag, making these antibodies valuable for diverse applications in CD14 research .
Comprehensive validation of CD14-biotin antibodies requires multiple complementary approaches. Flow cytometry should demonstrate strong staining of monocytes, weak staining of granulocytes, and minimal or no staining of lymphocytes, which serves as an excellent internal control system within peripheral blood samples. Blocking experiments using unlabeled anti-CD14 antibodies should significantly reduce binding of biotinylated antibodies. For antibodies compatible with Western blotting, validation should confirm a single band at the expected molecular weight (typically 40-55 kDa depending on glycosylation status). Cross-reactivity testing is essential when working with samples from multiple species, as many CD14 antibodies are species-specific. Comparison with alternative anti-CD14 clones targeting distinct epitopes (such as MEM-15, MEM-18, or 61D3) can further corroborate expression patterns. For definitive validation, immunoprecipitation with the antibody followed by mass spectrometry can confirm target identity .
Optimizing flow cytometry protocols with biotinylated CD14 antibodies requires attention to several critical parameters. First, perform thorough antibody titration to determine the optimal concentration, typically starting at ≤1 μg per 100 μL test volume containing 10^5-10^8 cells. Include appropriate blocking of Fc receptors using species-matched serum or commercial blocking reagents to reduce non-specific binding. For detection, select a streptavidin conjugate with a fluorophore suitable for your cytometer configuration and panel design, avoiding channels with high autofluorescence when possible. Essential controls include an isotype-matched biotinylated antibody as a negative control and streptavidin-only samples to assess background from the detection system. Leverage the differential expression of CD14 on blood cell populations as internal controls—monocytes (strong positive), granulocytes (weak positive), and lymphocytes (negative). When analyzing monocyte subsets, consider adjusting voltages to clearly distinguish CD14 expression levels across different populations. For multicolor panels, perform proper compensation and consider the potential spillover from the often bright signal generated by the biotin-streptavidin detection system .
Proper storage and handling of biotinylated CD14 antibodies is essential for maintaining their performance characteristics over time. Store antibodies at 4°C for short-term use (1-2 weeks) and at -20°C or -80°C for long-term storage, avoiding repeated freeze-thaw cycles by creating small working aliquots upon receipt. Most commercial biotinylated CD14 antibodies contain preservatives (e.g., 0.097% sodium azide) that help maintain stability, but note that sodium azide can inhibit enzymatic reactions and is toxic to cells in functional assays. When handling, allow antibodies to equilibrate to room temperature before opening to prevent condensation that could promote degradation. Use sterile technique and low protein-binding plasticware for dilutions. Regularly check for signs of degradation such as precipitates or aggregates. Follow manufacturer's recommendations for working dilutions in different applications, as optimal concentrations may vary substantially between flow cytometry, Western blotting, and ELISA. Document lot numbers, receipt dates, and aliquot usage to ensure experimental reproducibility .
Soluble CD14 (sCD14) can be precisely quantified using biotinylated CD14 antibodies through several methodological approaches, with sandwich ELISA being the most widely employed. In a typical sandwich ELISA configuration, a capture antibody against CD14 is immobilized on a microplate, followed by sample addition and detection with a biotinylated anti-CD14 antibody targeting a different epitope. This system can detect at least 2000-4000 pg/ml of recombinant human sCD14 protein when optimized. The biotin-streptavidin amplification system enhances sensitivity, particularly when using streptavidin-HRP conjugates with appropriate substrates. Western blot analysis using biotinylated antibodies provides an alternative approach that can distinguish between different forms of sCD14 based on molecular weight. When analyzing clinical samples, it's critical to include appropriate standards and controls, as elevated sCD14 levels are associated with sepsis and other inflammatory conditions. For multiplex analysis, bead-based assays can be developed using biotinylated CD14 antibodies alongside reagents for other soluble mediators, allowing simultaneous quantification of multiple biomarkers from limited sample volumes .
A robust control strategy is essential when using biotinylated CD14 antibodies in any immunoassay. Start with an isotype control—a biotinylated antibody of the same isotype (e.g., IgG1 for MEM-15 clone) with irrelevant specificity—to establish baseline non-specific binding. Include negative control samples (cells or tissues known not to express CD14, such as lymphocytes in flow cytometry) and positive control samples (monocytes, macrophages) to validate detection sensitivity. A secondary reagent-only control (samples treated only with the streptavidin detection reagent) is critical to assess background from the detection system itself. For blocking controls, pre-treat a set of samples with unlabeled anti-CD14 to demonstrate specificity through signal reduction. Include titration controls (a series of antibody dilutions) to confirm you're working at optimal concentration. For quantitative assays like ELISA, a standard curve using recombinant CD14 protein and inter-assay control samples are necessary to enable absolute quantification and normalize between experiments. When working with tissues or complex samples, consider adding controls for endogenous biotin blocking efficiency, especially for tissues known to contain high biotin levels .
CD14-biotin antibodies serve as powerful tools for dissecting monocyte/macrophage heterogeneity through multiple sophisticated approaches. In multi-parameter flow cytometry, biotinylated CD14 antibodies can be combined with antibodies against other lineage and functional markers (CD16, HLA-DR, CD86, CD163, etc.) to identify and quantify distinct subpopulations. The biotin-streptavidin system allows flexibility in fluorochrome selection to optimize panel design, particularly valuable for distinguishing classical (CD14++CD16-), intermediate (CD14++CD16+), and non-classical (CD14+CD16++) monocyte subsets. For cellular isolation, biotinylated CD14 antibodies coupled with streptavidin-coated magnetic beads enable purification of monocyte/macrophage populations based on CD14 expression levels for subsequent functional assays, transcriptomic analysis, or proteomics. In tissue contexts, immunohistochemistry or multiplexed immunofluorescence using biotinylated CD14 antibodies helps characterize tissue-resident macrophage populations in different anatomical locations. For cutting-edge applications, incorporating biotinylated CD14 antibodies into mass cytometry panels or single-cell RNA sequencing with protein detection (CITE-seq) allows correlation of CD14 protein expression with comprehensive phenotypic and transcriptomic profiles at single-cell resolution .
Resolving discrepancies in CD14 expression data requires systematic evaluation of both biological and technical factors. Standardize experimental protocols across comparisons, ensuring consistent sample preparation, antibody concentrations, incubation times, and detection methods. Cross-validate with multiple techniques (flow cytometry, Western blot, qPCR) to distinguish between technical artifacts and true biological variation. Employ multiple antibody clones (e.g., MEM-15, MEM-18, 61D3) that may recognize distinct epitopes differentially accessible depending on CD14's conformation, glycosylation state, or interaction with other molecules. Carefully evaluate whether discrepancies arise from differential detection of membrane-bound versus soluble CD14 forms. Consider biological variables including cell activation state, culture conditions, donor-to-donor variability, and disease states that can all profoundly alter CD14 expression patterns. Address technical variables by implementing quantitative standardization using calibration beads for flow cytometry, standard curves for ELISA, and normalization controls for Western blotting. Verify your findings against published literature, considering methodological differences that might explain inconsistencies. When discrepancies persist, collaborative cross-checking with different laboratories can identify environment-specific technical factors contributing to variability .
Biotinylated CD14 antibodies provide elegant solutions for investigating the LPS recognition complex through multiple advanced methodologies. For co-localization studies, combine biotinylated CD14 antibodies with streptavidin-conjugated fluorophores and antibodies against TLR4, MD-2 (LY96), or other signaling components to visualize receptor complex formation after LPS stimulation using confocal or super-resolution microscopy. Flow cytometry-based signaling studies can pair surface staining of CD14 (using biotinylated antibodies) with intracellular staining for phosphorylated signaling molecules (e.g., phospho-p38, phospho-NF-κB) to correlate CD14 expression levels with signaling pathway activation at the single-cell level. Leverage the biotin-streptavidin interaction for immunoprecipitation assays to isolate CD14-containing protein complexes after LPS stimulation, followed by identification of associated proteins through Western blotting or mass spectrometry. For dynamic studies, live-cell imaging using biotinylated Fab fragments of CD14 antibodies with fluorescent streptavidin allows tracking of receptor trafficking and clustering in real-time. These complementary approaches can reveal the composition, stoichiometry, and signaling consequences of CD14-containing receptor complexes, advancing our understanding of LPS recognition in innate immunity .
Successful multiplexing of biotinylated CD14 antibodies with other markers in flow cytometry requires careful attention to several technical considerations. First, address spectral overlap by selecting a streptavidin conjugate with minimal spillover into channels used for other markers in your panel, and prepare proper single-stained controls for accurate compensation. Follow strategic panel design principles by positioning CD14 detection in a channel appropriate for its expression level—bright channels are better suited for low-expression markers, while dimmer channels can be used for highly expressed markers like CD14 on monocytes. Implement a biotin blocking step (using unconjugated streptavidin followed by free biotin) before adding biotinylated antibodies to mitigate interference from endogenous biotin in samples. If using multiple biotinylated antibodies, sequential staining with intermediate blocking steps may be necessary. Verify antibody clone compatibility to ensure selected antibodies don't compete for overlapping epitopes or sterically hinder each other's binding. Re-titrate the biotinylated CD14 antibody in the context of the full panel, as optimal concentrations often differ from those determined in single-staining experiments. Finally, account for spreading error (signal spillover due to fluorescence intensity variations), which can be more pronounced with the high-intensity signals typically generated by biotin-streptavidin amplification systems .
Several factors can contribute to weak or absent signals when using biotinylated CD14 antibodies. Antibody degradation from improper storage (repeated freeze-thaw cycles) or use beyond expiration date is a common cause that can be addressed by using fresh aliquots from properly stored stocks. Epitope masking may occur when sample preparation methods like certain fixation protocols alter or mask the CD14 epitope recognized by the antibody—try alternative fixation methods or reduced fixation times. Low CD14 expression in the target population might reflect downregulation due to activation state, differentiation, or pathological conditions; comparing with known positive controls can help distinguish between technical and biological causes. Detection system issues such as degraded streptavidin conjugates or suboptimal concentrations require validation with alternative biotinylated antibodies. Endogenous biotin interference, particularly high in tissues like liver and kidney, can be mitigated through biotin blocking steps. Buffer incompatibilities, including high preservative concentrations, may interfere with biotin-streptavidin binding and can be addressed through buffer optimization. Systematic troubleshooting should include testing the antibody on known positive controls (e.g., fresh peripheral blood monocytes), verifying detection reagent activity, optimizing concentrations through careful titration, and adjusting incubation conditions .
High background when using biotinylated CD14 antibodies can be systematically addressed through several strategies. Implement endogenous biotin blocking by pre-incubating samples with free streptavidin followed by free biotin before adding biotinylated antibodies, which prevents streptavidin detection reagents from binding to naturally occurring biotin in biological samples. Reduce non-specific binding to Fc receptors by pre-incubating samples with serum from the same species as the secondary reagent or with commercial Fc receptor blocking reagents, particularly important when analyzing monocytes and macrophages which express high levels of Fc receptors. Optimize antibody concentration through careful titration experiments to determine the concentration that maximizes signal-to-noise ratio—excessive antibody often increases background more than specific signal. Improve washing procedures by increasing the number and volume of washes, and ensure thorough mixing during wash steps. Adjust blocking protocols by optimizing blocking buffer composition (e.g., BSA concentration, addition of normal serum) and duration. Use high-quality, filtered antibody preparations (e.g., "0.2 μm post-manufacturing filtered" as mentioned for some products) to reduce aggregates that contribute to background. For flow cytometry applications, incorporate steps to reduce cellular autofluorescence, which can be particularly problematic in fixed macrophages and tissue-derived samples .
Interpreting changes in CD14 expression across disease models requires contextual analysis that considers multiple factors. First, distinguish between membrane-bound CD14 (mCD14) and soluble CD14 (sCD14), as they may show different patterns—increased sCD14 with decreased mCD14 may indicate enhanced shedding, while elevated sCD14 without changes in mCD14 might reflect increased production by hepatocytes. Context-specific interpretation is essential, as increased soluble CD14 often correlates with disease severity in sepsis models and may serve as a biomarker for bacterial infection, while CD14 upregulation on monocytes/macrophages in atherosclerosis models may indicate enhanced foam cell formation and plaque development. Cell type-specific analysis is critical—changes on monocytes may indicate altered differentiation or activation states, while even small changes in the low baseline expression on granulocytes may be functionally significant. Temporal expression patterns provide important insights into disease progression: early upregulation may indicate roles in disease initiation, while late-stage changes may reflect compensatory mechanisms. Always correlate expression changes with functional readouts such as LPS responsiveness, phagocytic capacity, or inflammatory mediator production. Integrate CD14 data with other markers, including CD14/CD16 co-expression patterns for monocyte subset analysis and TLR4/MD-2 expression for LPS recognition complex status .
Multiple factors influence the detection sensitivity of biotinylated CD14 antibodies across different applications. The biotin:antibody ratio is critical—insufficient biotinylation reduces detection sensitivity, while excessive biotinylation can impair antibody binding affinity or create steric hindrance. Detection system selection dramatically affects sensitivity, with amplification systems (e.g., streptavidin-poly-HRP) often providing 10-100 fold enhancement over conventional systems. Epitope accessibility varies with sample preparation methods—some fixation protocols may mask the CD14 epitope, particularly if the epitope is conformational or near the membrane. Buffer composition influences antibody-antigen interactions; optimal buffers typically contain carrier proteins (BSA) and appropriate salt concentrations to minimize non-specific binding while maximizing specific signal. Incubation conditions affect binding kinetics—generally, longer incubation times at 4°C favor equilibrium binding and can improve sensitivity for low-abundance targets. Instrument sensitivity in flow cytometry applications (PMT voltage settings, laser power) or detection limits in Western blotting and ELISA affect the minimal detectable signal. Sample quality issues including protein degradation, excessive cell death, or interfering substances can reduce target availability or accessibility. Systematic optimization of these parameters can significantly enhance detection sensitivity, potentially enabling identification of CD14 on cell populations with low expression levels .
Biotinylated CD14 antibodies are enabling sophisticated investigations in sepsis research through multiple innovative applications. For monitoring monocyte function, flow cytometric analysis using biotinylated CD14 antibodies in combination with HLA-DR and other markers helps track monocyte deactivation—a hallmark of immunosuppression in sepsis—enabling correlation of CD14 expression levels with functional impairment and disease outcomes. High-dimensional phenotyping incorporating biotinylated CD14 antibodies into mass cytometry or spectral cytometry panels allows comprehensive immune profiling of sepsis patients, revealing previously unrecognized cellular subsets and activation states associated with disease progression or resolution. Dual-detection assays using differently labeled anti-CD14 antibodies (including biotinylated variants) can simultaneously quantify membrane-bound and soluble CD14, providing insights into the dynamics of CD14 shedding during sepsis, where elevated sCD14 levels often correlate with disease severity and may serve as biomarkers. For tissue analysis, immunohistochemistry or multiplexed immunofluorescence using biotinylated CD14 antibodies enables characterization of tissue macrophage phenotypes in post-mortem samples from sepsis patients. These applications contribute to a more nuanced understanding of CD14's role in sepsis pathophysiology and may inform the development of novel diagnostic and therapeutic approaches .
CD14 plays multifaceted roles in atherosclerosis, and biotinylated CD14 antibodies provide powerful tools for investigating these mechanisms. As a mediator of lipoprotein interactions, CD14 binds electronegative LDL (LDL(-)) and mediates pro-inflammatory cytokine release induced by LDL(-), contributing to foam cell formation and plaque development. Through its function as a coreceptor for TLR4 and TLR2, CD14 facilitates recognition of endogenous damage-associated molecular patterns (DAMPs) in the vessel wall, promoting inflammatory responses that drive atherosclerosis progression. Biotinylated CD14 antibodies enable plaque composition analysis through immunohistochemistry or multiplexed immunofluorescence to characterize macrophage populations within atherosclerotic lesions, identify their distribution in different plaque regions, and correlate CD14 expression with markers of inflammation or stability. Flow cytometric phenotyping using biotinylated CD14 antibodies allows detailed characterization of monocyte subsets in atherosclerosis models, particularly tracking CD14+CD16- (classical) versus CD14+CD16+ (intermediate/non-classical) monocytes, which have different propensities for recruitment to atherosclerotic plaques. For functional studies, isolation of CD14+ cells using biotinylated antibodies and streptavidin beads enables ex vivo assessment of foam cell formation capacity, analysis of inflammatory responses to modified lipoproteins, and gene expression profiling of plaque-derived CD14+ cells .
Biotinylated CD14 antibodies are facilitating novel investigations in the rapidly evolving field of extracellular vesicle (EV) research. For CD14+ EV isolation and characterization, biotinylated CD14 antibodies coupled to streptavidin-coated magnetic beads enable selective capture of CD14+ EVs from complex biological fluids, while flow cytometric analysis using biotinylated CD14 antibodies with streptavidin-fluorophores allows high-sensitivity detection of CD14 on individual EVs using specialized flow cytometers. Functional studies of CD14+ EVs explore their potential roles as intercellular communication mediators, using biotinylated antibodies to track the transfer of CD14 via EVs to recipient cells that normally don't express CD14, potentially conferring LPS responsiveness to these cells. In disease-specific applications, quantifying CD14+ EVs in plasma serves as a potential early biomarker of sepsis or indicator of treatment response, while monitoring changes in CD14+ EV profiles during inflammatory disease progression or following therapeutic intervention provides insights into disease mechanisms. Technological innovations pair biotinylated CD14 antibodies with nanoscale flow cytometry for analyzing individual vesicles below 200 nm or with super-resolution microscopy for visualizing CD14 distribution on individual EVs. These emerging applications highlight the versatility of biotinylated CD14 antibodies in advancing our understanding of extracellular vesicle biology in both physiological and pathological contexts .
Biotinylated CD14 antibodies offer sophisticated approaches for investigating TLR signaling complexes through multiple complementary techniques. Multi-color super-resolution microscopy combining biotinylated CD14 antibodies with directly labeled antibodies against TLR4, MD-2, and other signaling components enables nanoscale visualization of receptor clusters before and after ligand stimulation. Proximity ligation assays utilize biotinylated CD14 antibodies with antibodies against TLR4/TLR2 to provide quantitative assessment of receptor proximity and complex formation with single-molecule sensitivity. For biochemical characterization, co-immunoprecipitation with biotinylated CD14 antibodies and streptavidin beads offers efficient pull-down of CD14-associated complexes for Western blot or mass spectrometry analysis. Live-cell imaging using biotinylated Fab fragments of CD14 antibodies with fluorescent streptavidin allows tracking of receptor trafficking and clustering in real-time. Specialized approaches like lipid raft isolation using biotinylated CD14 antibodies can track partitioning of CD14 and associated TLRs into membrane microdomains, which is critical for signaling. Advanced single-cell approaches incorporate biotinylated CD14 antibodies into mass cytometry panels with antibodies against signaling pathway components to correlate receptor expression with activation states at single-cell resolution. These methodologies provide unprecedented insights into the composition, dynamics, and signaling consequences of CD14-containing receptor complexes in innate immunity .
Cluster of Differentiation 14 (CD14) is a protein primarily expressed on the surface of monocytes and macrophages, which are key players in the innate immune system . CD14 acts as a co-receptor along with Toll-like receptor 4 (TLR4) and MD-2 for the detection of bacterial lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria . This protein exists in two forms: a membrane-bound form (mCD14) and a soluble form (sCD14) .
CD14 plays a crucial role in the immune response by recognizing and binding to LPS, which triggers a cascade of signaling events leading to the activation of the immune system . It also recognizes other pathogen-associated molecular patterns (PAMPs) such as lipoteichoic acid from Gram-positive bacteria .
Mouse Anti-Human Biotin antibodies are monoclonal antibodies produced in mice that are specific to human antigens and conjugated with biotin . Biotin, also known as Vitamin B7, is a coenzyme that is widely used in molecular biology due to its strong affinity for avidin and streptavidin, which allows for easy detection and purification of biotinylated molecules .
These antibodies are commonly used in various immunoassays, including:
The biotinylation of antibodies enhances their utility in these assays by allowing for the use of avidin or streptavidin conjugates for detection, which can be labeled with enzymes, fluorophores, or other markers .
The combination of CD14 and Mouse Anti-Human Biotin antibodies is particularly useful in research focused on the immune response to bacterial infections. By using biotinylated antibodies specific to human CD14, researchers can: