CD14 is a 53–55 kDa glycoprotein expressed on monocytes, macrophages, and neutrophils. It acts as a co-receptor for Toll-like receptors (TLRs), facilitating recognition of pathogen-associated molecular patterns (PAMPs) like lipopolysaccharide (LPS) . Overactivation of CD14 can trigger hyperinflammatory responses, contributing to conditions such as sepsis, acute respiratory distress syndrome (ARDS), and amyotrophic lateral sclerosis (ALS) .
Several CD14 mAbs have been developed, each with distinct properties:
CD14 mAbs function via:
Receptor Blockade: IC14 binds membrane-bound (mCD14) and soluble (sCD14) forms, preventing LPS-CD14 interactions and downstream proinflammatory signaling .
Immunomodulation: In ALS, IC14 enhances regulatory T-cell (Treg) activity, potentially slowing disease progression .
Pathogen Neutralization: MEM-15 inhibits LPS-induced cytokine release, mitigating septic shock .
Design: Phase 2 trial (NCT04391309) testing IC14 + remdesivir in 300–350 hospitalized patients .
Outcomes: No significant reduction in recovery time, but trend toward lower mortality (data pending peer review) .
Expanded Access Protocol: 17 patients received IC14 for ≤103 weeks.
Safety: Mild adverse events; 96–104% monocyte CD14 receptor occupancy achieved .
Rabbit Model: Anti-CD14 mAb administered post-LPS exposure reduced mortality by 80%, outperforming anti-TNF therapies .
Flow Cytometry: 61D3 and MEM-15 enable immune cell profiling .
Western Blot: CAB19011 (rabbit mAb) detects CD14 in human samples .
Inflammation Studies: MAB3832 elucidates CD14’s role in TLR signaling .
This product is a mouse monoclonal antibody targeting human CD14 (amino acids 20-345), encompassing the mature extracellular domain in its membrane-bound GPI-anchored form. The IgG1 isotype antibody is purified by protein A chromatography, achieving >95% purity. It exhibits cross-reactivity with CD14 from human, mouse, and rabbit. This antibody is suitable for ELISA, Western blotting, flow cytometry, immunofluorescence, and immunohistochemistry. It is a valuable tool for immunological research, particularly in characterizing disease-associated immune pathways and pro-inflammatory responses to pathogens. Human CD14, a monocyte differentiation antigen, plays a critical role in activating the innate immune response. Expressed on the surface of monocytes and most tissue macrophages, CD14 regulates immune responses to bacterial lipopolysaccharide (LPS), and also mediates cellular and immune responses to diacylated and triacylated lipopeptides, and electronegative low-density lipoprotein (LDL-).
CD14 serves as a coreceptor for bacterial lipopolysaccharide (LPS). In conjunction with lipopolysaccharide-binding protein (LBP), it binds monomeric LPS and delivers it to the Toll-like receptor 4 (TLR4)/MD-2 complex, thereby initiating the innate immune response to bacterial LPS. This process involves MyD88, TIRAP, and TRAF6, leading to NF-κB activation, cytokine secretion, and the subsequent inflammatory response. CD14 also functions as a coreceptor for the TLR2/TLR6 heterodimer in response to diacylated lipopeptides and the TLR2/TLR1 heterodimer in response to triacylated lipopeptides. These complexes trigger signaling from the cell surface and are subsequently trafficked to the Golgi apparatus via a lipid-raft dependent pathway. Furthermore, CD14 binds electronegative LDL (LDL-) and mediates the release of cytokines induced by LDL-.
Applications : WB
Sample type: cells
Review: CD14 protein levels were determined by Western blot analysis and were quantified using the ImageJ program.
CD14 is a 53-55 kDa glycosylphosphatidylinositol (GPI)-linked glycoprotein predominantly expressed on the surface of mature monocytes, macrophages, and neutrophils. It functions as a pattern recognition receptor, particularly as a multifunctional lipopolysaccharide (LPS) receptor. CD14 exists in two forms: a membrane-bound form (mCD14) and a soluble form (sCD14) that circulates in serum. The soluble form results from both secretion and enzymatic cleavage of the GPI-anchored form. CD14's primary function involves binding LPS in a reaction catalyzed by LPS-binding protein (LBP), an acute phase serum protein, facilitating the recognition of bacterial components by the immune system. Additionally, CD14 has been shown to associate with Toll-Like Receptor 4 (TLR4) to participate in signaling and cellular response to bacterial LPS, making it a critical component in innate immunity and inflammatory responses.
CD14 monoclonal antibodies are employed in numerous research applications, with the most common being:
Flow cytometric analysis: CD14 antibodies are widely used to identify and characterize monocyte populations in peripheral blood and other tissues. This application typically requires careful titration of the antibody, with recommended concentrations of ≤0.5 μg per test for human samples.
Immunohistochemistry: Anti-CD14 antibodies can be used for staining formalin-fixed paraffin-embedded (FFPE) tissue sections, typically requiring antigen retrieval techniques. For optimal results, concentrations of ≤20 μg/mL are recommended for human tissue samples.
Western blot analysis: CD14 antibodies can detect CD14 protein in cell lysates, showing specific bands at approximately 55 kDa, particularly in human peripheral blood mononuclear cells (PBMCs).
Functional studies: Certain anti-CD14 antibodies can be used in functional assays to block CD14 activity, providing insights into its role in inflammatory pathways and responses to bacterial components.
The selection of an appropriate CD14 monoclonal antibody clone depends on several factors:
Species specificity: Different clones target CD14 from different species. For example, clone 61D3 reacts with human CD14, while Sa2-8 targets mouse CD14.
Application compatibility: Some clones are optimized for specific applications. For instance, clone 61D3 has been validated for flow cytometry, immunohistochemistry, and functional studies, while functional assays specifically recommend using the Functional Grade Purified 61D3 (Product #16-0149).
Functional properties: Certain clones possess antagonistic properties. For example, Sa2-8 demonstrates weak antagonistic activity in NF-kappaB activation or TNF-alpha production with LPS stimulation, making it suitable for functional studies examining these pathways.
Epitope recognition: Different clones recognize different epitopes on CD14, which may affect their suitability for specific research questions, particularly when studying structural variations or conformational changes in CD14.
For optimal results, researchers should carefully titrate the selected antibody for their specific application and experimental conditions.
When employing CD14 monoclonal antibodies in flow cytometric analysis, researchers should consider several critical factors to ensure reliable and reproducible results:
Antibody titration: Thorough antibody titration is essential for determining the optimal concentration. For human samples, ≤0.5 μg per test is typically recommended for clones like 61D3, while mouse-specific clones like Sa2-8 may require up to 1 μg per test.
Cell number optimization: Cell numbers should be empirically determined but typically range from 10^5 to 10^8 cells per test in a final volume of 100 μL.
Fluorophore selection: The choice of fluorophore should align with the available laser configuration of the flow cytometer. For instance, FITC-conjugated antibodies (excitation: 488 nm; emission: 520 nm) require blue laser compatibility.
Gating strategy: When analyzing peripheral blood, appropriate gating strategies are crucial to distinguish CD14-positive monocytes from other cell populations.
Controls: Proper isotype controls should be included to account for non-specific binding and facilitate accurate interpretation of results.
Sample preparation: Fresh samples typically yield better results, but if fixed samples are used, the fixation method may affect epitope recognition and should be validated.
CD14 monoclonal antibodies serve as valuable tools for investigating CD14's role in sepsis-induced coagulopathy through several methodological approaches:
In vivo inhibition studies: Anti-CD14 antibodies can be administered in animal models of sepsis to assess their impact on coagulation parameters. Research has demonstrated that CD14 blockade significantly reduces factor XI activation, factor VIIa generation, and thrombin-antithrombin (TAT) complex formation, indicating inhibition of both the activation and amplification phases of coagulation.
Tissue factor expression analysis: CD14 inhibition significantly reduces E. coli-induced tissue factor (TF) mRNA expression in circulating blood cells and lung tissue. Researchers can use CD14 monoclonal antibodies to block CD14 function and subsequently measure TF expression to elucidate the relationship between CD14 signaling and TF upregulation.
Fibrinolysis assessment: CD14 blockade substantially affects fibrinolysis parameters, leading to decreased plasma PAI-1 (by ~90%), increased tissue plasminogen activator (tPA) levels, enhanced plasmin generation (three-fold increase in PAP complexes), and increased fibrin degradation (two-fold increase in D-dimer). These parameters can be measured following CD14 inhibition to understand its role in fibrinolytic balance during sepsis.
Endothelial cell integrity evaluation: Anti-CD14 treatment reduces the formation of complexes between activated protein C and its inhibitor α1-antitrypsin, correlating with decreased thrombin generation and better preservation of endothelial cell integrity.
Successful immunohistochemical staining with CD14 monoclonal antibodies requires attention to several technical aspects:
Antigen retrieval: For formalin-fixed paraffin-embedded (FFPE) tissues, effective antigen retrieval is crucial. Low pH antigen retrieval methods are recommended for optimal CD14 detection. Specific protocols like VisUCyte Antigen Retrieval Reagent-Basic have been successfully employed.
Antibody concentration: CD14 antibodies for IHC typically require optimization, with concentrations of ≤20 μg/mL for human tissues. Specific protocols have demonstrated successful staining at 5 μg/mL when incubated for 1 hour at room temperature.
Detection systems: Secondary antibody selection and detection systems significantly impact staining quality. HRP-conjugated detection systems, such as the Anti-Mouse IgG VisUCyte HRP Polymer Antibody, have been successfully used with CD14 antibodies like clone MAB3832.
Tissue-specific considerations: Different tissues may require different protocols. For instance, human tonsil sections have shown specific CD14 staining localized to cell surfaces in lymphocytes using clone MAB3832.
Controls: Appropriate positive and negative controls are essential to validate staining specificity and optimize protocols.
Counterstaining: Hematoxylin counterstaining provides contrast for visualizing tissue architecture alongside CD14-positive cells.
Recent research has explored the therapeutic potential of CD14 monoclonal antibodies in various inflammatory conditions, particularly focusing on severe infectious and inflammatory diseases:
COVID-19 respiratory disease: A Phase 2 clinical trial (COVID-19 anti-CD14 Treatment Trial, CaTT) by the National Institute of Allergy and Infectious Diseases has investigated the safety and efficacy of IC14, an investigational monoclonal antibody targeting CD14, for treating hospitalized COVID-19 patients with respiratory disease and low blood oxygen. The trial enrolled 300-350 patients aged 18 years or older across multiple sites.
Mechanism of action in COVID-19: The therapeutic rationale stems from CD14's role in potentially overamplifying immune responses during SARS-CoV-2 infection. By blocking CD14 during early disease stages, IC14 aims to temper harmful inflammatory responses, limit associated tissue damage, and improve clinical outcomes by preventing cytokine storms that can lead to acute respiratory distress syndrome.
Sepsis-induced coagulopathy: Research has demonstrated that CD14 inhibition improves survival and attenuates thromboinflammation in sepsis models. Anti-CD14 treatment reduces coagulation activation, particularly through the extrinsic pathway, by decreasing tissue factor expression. Moreover, it enhances fibrinolysis by maintaining higher tPA levels, significantly reducing PAI-1, and increasing plasmin generation and fibrin degradation.
Researchers employ various experimental models and assays to evaluate CD14-targeting therapeutic strategies:
In vivo sepsis models: Animal models, particularly non-human primates like baboons, have been used to study the effects of anti-CD14 antibodies on sepsis-induced coagulopathy, inflammation, organ dysfunction, and mortality. These models typically involve challenging the animals with E. coli and subsequently administering anti-CD14 antibodies.
Coagulation assays: Multiple assays are used to evaluate the impact of CD14 inhibition on coagulation parameters:
Fibrinolysis assessment: Researchers measure various parameters to evaluate the effect of CD14 inhibition on fibrinolysis:
Gene expression analysis: Quantitative PCR is used to measure tissue factor mRNA expression in circulating blood cells and tissues, providing insights into how CD14 inhibition affects procoagulant responses.
Clinical trials: Human clinical trials, such as the CaTT trial for COVID-19, evaluate safety parameters, efficacy endpoints (including mortality, ventilator-free days, and clinical status improvements), and biomarkers of inflammation and coagulation.
Researchers frequently encounter several challenges when working with CD14 monoclonal antibodies, each requiring specific troubleshooting approaches:
Variable expression levels: CD14 expression can vary significantly between different cell populations and under different physiological conditions. Solution: Include appropriate positive controls (such as monocyte populations) and carefully validate CD14 detection in specific experimental contexts.
Non-specific binding: Some CD14 antibodies may exhibit cross-reactivity or non-specific binding. Solution: Optimize antibody concentration through careful titration, use appropriate blocking reagents, and include proper isotype controls to distinguish specific from non-specific signals.
Epitope masking: Certain sample preparation methods may mask the CD14 epitope. Solution: For fixed samples, optimize fixation protocols and consider using antigen retrieval methods, particularly for FFPE tissues.
Batch-to-batch variability: Antibody performance may vary between lots. Solution: Test new antibody lots against previous lots using standardized samples and protocols to ensure consistent performance.
Fluorophore selection in multicolor flow cytometry: CD14 is often used in panels with other markers, and spectral overlap can complicate analysis. Solution: Choose fluorophores that minimize spectral overlap with other markers in the panel and perform proper compensation.
To ensure reliable and reproducible results with CD14 monoclonal antibodies, researchers should implement several quality control measures:
Antibody validation: Verify antibody specificity using positive and negative control samples. For human samples, peripheral blood monocytes serve as excellent positive controls.
Purity assessment: Commercial antibodies should meet specific purity criteria. For instance, the 61D3 monoclonal antibody is reported to have >90% purity as determined by SDS-PAGE and <10% aggregation as assessed by HPLC.
Filtration verification: Ensure antibodies have undergone appropriate filtration (e.g., 0.2 μm post-manufacturing filtration) to remove particulates and aggregates that could affect performance.
Application-specific controls:
For flow cytometry: Include appropriate isotype controls, fluorescence minus one (FMO) controls, and single-stain controls for compensation.
For Western blot: Include molecular weight markers and positive control lysates (e.g., human PBMC lysates).
For IHC: Include positive control tissues known to express CD14 (e.g., human tonsil) and negative control sections without primary antibody.
Functional verification: For antibodies intended for functional studies, verify their blocking or stimulating capacity in appropriate assay systems before conducting extensive experiments.
Recent research utilizing CD14 monoclonal antibodies has expanded our understanding of inflammatory processes in various conditions:
COVID-19 pathophysiology: Studies investigating the role of CD14 in COVID-19 have revealed that CD14 may contribute to the hyperactive inflammatory response and cytokine storm observed in severe cases. Anti-CD14 monoclonal antibody therapy (IC14) has entered clinical trials as a potential treatment approach, highlighting CD14's significance in viral respiratory diseases.
Thromboinflammation: Research has demonstrated that CD14 plays a crucial role in the interplay between inflammation and coagulation. CD14 inhibition reduces tissue factor expression, factor VIIa generation, and thrombin formation while enhancing fibrinolysis, suggesting that CD14 contributes to the prothrombotic state in inflammatory conditions.
Endothelial dysfunction: CD14 blockade improves endothelial cell integrity during inflammatory states, as evidenced by reduced formation of complexes between activated protein C and its inhibitor. This suggests CD14's involvement in endothelial damage during inflammation.
Pattern recognition beyond LPS: While CD14 is primarily known as an LPS receptor, ongoing research is exploring its role in recognizing other pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), suggesting broader implications in sterile inflammation and tissue injury responses.
Several promising research directions are emerging in the field of CD14 monoclonal antibodies:
Therapeutic development: Building on the clinical trials of IC14 for COVID-19, researchers are exploring the potential of CD14-targeted therapies for other conditions characterized by dysregulated inflammation, including sepsis, acute respiratory distress syndrome, and autoimmune diseases.
Biomarker development: CD14 expression patterns and soluble CD14 levels are being investigated as potential biomarkers for disease severity, progression, and treatment response in various inflammatory conditions.
Combination therapies: Research is exploring the synergistic effects of combining CD14 blockade with other immunomodulatory approaches, such as targeting toll-like receptors or cytokine pathways, to achieve more comprehensive control of inflammatory responses.
Novel antibody engineering: Development of engineered anti-CD14 antibodies with enhanced specificity, improved tissue penetration, or dual targeting capabilities could expand therapeutic applications.
Precision medicine approaches: Identifying patient subpopulations that would benefit most from CD14-targeted interventions based on biomarker profiles or genetic factors could optimize therapeutic outcomes and minimize adverse effects.