CD14 Antibody

CD14, Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of CD14

CD14 is a 55 kDa glycosylphosphatidylinositol (GPI)-anchored glycoprotein that functions as a pattern recognition receptor (PRR) in innate immunity . It binds pathogen-associated molecular patterns (PAMPs), such as bacterial lipopolysaccharide (LPS), and damage-associated molecular patterns (DAMPs), including oxidized lipids, to activate Toll-like receptors (TLRs) like TLR4 and TLR2 . CD14 exists in two forms:

  • Membrane-bound CD14 (mCD14): Expressed on monocytes, macrophages, neutrophils, and some epithelial cells .

  • Soluble CD14 (sCD14): Circulates in plasma and can transfer ligands to cells lacking mCD14, such as endothelial cells .

CD14 antibodies, such as IC14 (atibuclimab), are monoclonal antibodies designed to block CD14-mediated inflammatory signaling .

Mechanism of Action of CD14 Antibodies

CD14 antibodies inhibit ligand binding to CD14, thereby disrupting downstream pro-inflammatory pathways:

  • TLR4 Signaling Blockade: CD14 facilitates LPS transfer to the TLR4-MD-2 complex, triggering NF-κB and IRF3 activation . Anti-CD14 antibodies prevent this interaction, reducing cytokine production (e.g., TNF-α, IL-6) .

  • Phagocytosis Modulation: CD14 blockade minimally affects bacterial phagocytosis by monocytes and granulocytes, preserving host defense .

  • Soluble CD14 Neutralization: Antibodies like IC14 bind sCD14, limiting its role in amplifying inflammation in non-immune cells .

Table 1: Clinical Trials Involving CD14 Antibodies

Study PhaseConditionAntibodyKey OutcomesSource
Phase 1SepsisIC14Reduced LPS-induced TNF-α (90%) and IL-6 (85%) in healthy volunteers .
Phase 2COVID-19 ARDSIC14Reduced BAL neutrophil counts and plasma cytokines; ongoing trial .
Phase 1/2ALSIC14Safe up to 103 weeks; 80% CD14 receptor occupancy achieved .

Table 2: In Vitro/Ex Vivo Studies

ModelInterventionEffectSource
Human monocytesAnti-CD14 (18E12)Inhibited TNF production by 40–65% during H. influenzae infection .
Mouse macrophagesCD14 knockoutResistant to LPS-induced septic shock .
Human PBMCsIC14Reduced TLR4 endocytosis and TRIF-dependent IFN-β production .

Therapeutic Applications

  • Sepsis: IC14 reduced cytokine storms in primates and humans, improving survival in preclinical models .

  • COVID-19: By targeting CD14, IC14 mitigates hyperinflammation in severe SARS-CoV-2 cases, as sCD14 levels correlate with disease severity .

  • Neurodegenerative Diseases: In ALS, IC14 showed safety in long-term use (103 weeks) without impairing phagocytosis .

Comparative Analysis with Other Antibodies

TargetAntibodyEpitope SpecificityClinical Impact
CD14IC14Extracellular domainBlocks LPS/TLR4 signaling
TLR4TAK-242Intracellular domainFailed in sepsis trials due to toxicity .
CD11bLeukadherinIntegrin-binding siteReduces leukocyte adhesion .

Future Directions

  • Combination Therapies: Pairing IC14 with antivirals (e.g., remdesivir) for synergistic effects in COVID-19 .

  • Chronic Inflammation: Exploring CD14’s role in metabolic diseases (e.g., atherosclerosis) and autoimmune disorders .

  • Biomarker Development: sCD14 levels as a prognostic marker for ARDS and sepsis .

Product Specs

Introduction
CD14, also known as the lipopolysaccharide (LPS) receptor, is primarily found on the surface of monocytes and macrophages. Neutrophils exhibit lower levels of CD14 expression. This protein is attached to the cell membrane via a glycosylphosphatidylinositol (GPI) anchor. CD14 functions as a high-affinity receptor for complexes formed by LPS and LPS binding protein (LBP). In its soluble form, CD14 can act as an agonist for LPS at physiological concentrations, while at higher concentrations, it exhibits an antagonistic effect on LPS-induced cell activation. Furthermore, CD14 has been observed to interact with apoptotic cells.
Formulation
The antibody is provided as a lyophilized powder. Upon reconstitution, the concentration will be 1 mg/ml in a phosphate-buffered saline (PBS) solution.
Storage Procedures
The lyophilized antibody should be stored at 4°C. After reconstitution, it is recommended to use the antibody within one month. If longer storage is required, aliquoting and storage at -20°C is advised.
Solubility
To reconstitute the antibody, add deionized water (H2O) to the vial. Mix gently by inverting or swirling, ensuring the solution makes contact with the sides of the vial. Allow 30-60 seconds for the antibody to fully dissolve before use.
Applications
This antibody can be employed for blocking experiments and cell staining procedures. For staining applications, use 10 µl of antibody per 1,000,000 cells. The optimal antibody concentration for blocking LPS binding should be determined empirically by the researcher.
Available Conjugates
In addition to the unconjugated form, this antibody is also available with biotin and fluorescein isothiocyanate (FITC) conjugates. When utilizing biotin- or FITC-conjugated antibodies for staining, it is recommended to use 5-10 µl of antibody for every 1,000,000 cells.
Synonyms
Monocyte differentiation antigen CD14, Myeloid cell-specific leucine-rich glycoprotein.
Purification Method
Ion exchange column.
Type
Mouse Anti Human Monoclonal.
Clone
hCD14.
Immunogen
Purified human B Cells Ig SubclassMouse IgG1.

Q&A

What is CD14 and why is it a significant target for antibody development?

CD14 is a myeloid differentiation antigen expressed primarily on peripheral blood monocytes, dendritic cells, and macrophages. It functions as a key regulator of inflammatory responses to gram-negative bacteria, oxidative burst, and septic shock . The significance of CD14 as an antibody target stems from its critical role in the immune response pathway. CD14 helps immune cells recognize pathogens and injured or dying cells, alerting the immune system to danger and prompting it to respond .

During infections like SARS-CoV-2, CD14 can overamplify the later stages of the immune response, potentially leading to hyperactive inflammatory responses and cytokine storms . These excessive immune reactions can cause severe tissue damage, particularly in the lungs, resulting in acute respiratory distress syndrome and respiratory failure. The inflammatory cascade initiated through CD14 signaling makes it an attractive target for therapeutic intervention in multiple inflammatory conditions.

Research suggests that during SARS-CoV-2 infection in the lungs, CD14 overamplifies the immune response to the virus, potentially leading to a hyperinflammatory state and cytokine storm. By developing antibodies that can modulate CD14 activity, researchers aim to control excessive inflammation while preserving essential immune functions .

How can researchers validate the specificity of a CD14 antibody?

Validating CD14 antibody specificity requires a systematic, multi-platform approach to ensure reliable experimental results. A comprehensive validation protocol should include the following methodological steps:

  • Flow cytometry validation: Test the antibody against peripheral blood cells with known CD14 expression patterns. Monocytes should show high CD14 expression, while lymphocytes should be negative or show very low expression. Include CD14-negative cell lines (e.g., NALM-6) as negative controls. Compare reactivity patterns with previously validated anti-CD14 antibodies .

  • Immunoprecipitation and Western blot analysis: Prepare whole cell lysates from fresh peripheral blood leukocytes and run alongside red blood cells with platelets as a negative control. The CD14 antibody should precipitate a protein of approximately 40-55 kDa (variation due to glycosylation) from leukocytes but not from the negative control samples .

  • Cross-validation with established antibodies: Compare your antibody's binding pattern with previously validated anti-CD14 antibodies (such as clone biG 10) that have known cross-reactivity and specificity. For instance, in one study, immunoprecipitation with a validated anti-human CD14 antibody (clone biG 10) was used to confirm the specificity of a new anti-CD14 antibody .

  • Tissue staining verification: Perform immunohistochemistry on tissues known to contain CD14-expressing cells, such as liver sinusoids and tonsil, using appropriate controls. Specific staining should be localized to expected anatomical locations, such as sinusoids in liver tissue .

  • Functional blocking assays: Verify that the antibody can block LPS binding to CD14, which can be measured by flow cytometry or functional assays. This provides not only specificity validation but also confirms the antibody's functional relevance .

By implementing this multi-faceted validation approach, researchers can confidently establish the specificity of their CD14 antibody and proceed with experimental applications.

What detection methods can be employed for CD14 antibody application in research?

CD14 antibodies can be utilized across multiple detection platforms, each requiring specific methodological considerations:

Flow Cytometric Analysis:

  • Sample preparation: Isolate peripheral blood mononuclear cells (PBMCs) using density gradient centrifugation

  • Cell concentration: Approximately 5×10^5 cells per test tube

  • Antibody incubation: Primary incubation with anti-CD14 antibody followed by detection with fluorescently-labeled secondary antibodies

  • Controls: Include isotype controls and CD14-negative cell lines (e.g., NALM-6)

  • Analysis: Use flow cytometry software (e.g., CELLQUEST) to analyze fluorescent signals

Western Blot Analysis:

  • Sample preparation: Prepare cell lysates from monocytic cell lines (e.g., THP-1)

  • Treatment conditions: For enhanced detection, cells can be treated with PMA (200 nM for 24 hours) followed by LPS (10 μg/mL for 3 hours)

  • Membrane type: PVDF membrane recommended

  • Antibody concentration: Approximately 1 μg/mL

  • Detection: Use HRP-conjugated secondary antibodies and appropriate detection reagents

  • Expected band size: Approximately 55 kDa under non-reducing conditions

Immunohistochemistry:

  • Tissue preparation: Immersion fixed paraffin-embedded sections

  • Epitope retrieval: Heat-induced epitope retrieval using basic retrieval reagents

  • Antibody concentration: 3 μg/mL for 1 hour at room temperature

  • Detection system: HRP Polymer detection systems

  • Expected staining: Sinusoids in liver tissue, macrophages in tonsil

Immunoprecipitation:

  • Sample source: Whole cell lysates from peripheral blood leukocytes

  • Controls: Red blood cells with platelets as negative controls

  • Detection method: Following immunoprecipitation, Western blot analysis can confirm the expected molecular weight of CD14

Each detection method offers unique advantages for studying CD14 expression and function. Flow cytometry provides quantitative analysis of CD14 on cell surfaces, Western blotting confirms molecular weight and protein integrity, immunohistochemistry reveals tissue distribution patterns, and immunoprecipitation allows for studying protein interactions. Selecting the appropriate method depends on the specific research question and available resources.

How can researchers design chimeric anti-CD14 antibodies to reduce immunogenicity?

Designing chimeric anti-CD14 antibodies requires a sophisticated approach to maintain binding specificity while reducing immunogenicity. Based on successful examples in research, the following methodology is recommended:

  • Antibody fragment selection: Start with a well-characterized murine anti-CD14 antibody with strong binding affinity and specificity. The single-chain fragment variable (scFv) portion, containing the VH and VL domains, should be retained as it determines the antigen-binding specificity .

  • Human constant region incorporation: Replace the murine constant regions with human IgG1 Fc regions, specifically the hinge, CH2, and CH3 domains. This chimeric structure (mouse variable regions + human constant regions) significantly reduces immunogenicity while maintaining antigen binding .

  • Expression vector construction: Design a universal vector system that allows for efficient cloning and expression, which typically includes:

    • A strong promoter (e.g., CMV promoter)

    • Signal peptide sequence for secretion

    • Restriction sites flanking the scFv and Fc regions

    • Selection markers (e.g., neomycin resistance)

  • Expression system selection: Chinese hamster ovary (CHO) cells are preferred for expression due to their ability to perform proper protein folding and post-translational modifications essential for antibody functionality .

  • Purification strategy: Implement a two-step purification process:

    • Protein A or G affinity chromatography (utilizes binding to Fc region)

    • Size exclusion chromatography (ensures purity and removes aggregates)

  • Validation of chimeric antibody:

    • Perform SDS-PAGE to confirm the expected molecular weight (~150 kDa)

    • Verify antigen-binding capacity using flow cytometry

    • Compare binding affinity with the parental murine antibody

    • Assess functional activity, such as blocking LPS binding to CD14

In one successful example, researchers constructed a chimeric antibody named Hm2F9 composed of anti-CD14 scFv and the Fc region of human IgG1. This antibody retained strong specific binding to CD14 with activity comparable to its murine parent (99.07% vs. 98.86% positive cells) while exhibiting the ability to block LPS binding to CD14 with high efficiency (reducing positive cells from 98.37% to 1.35%) .

What are the critical considerations in developing anti-CD14 antibodies for therapeutic applications?

Developing anti-CD14 antibodies for therapeutic applications involves multiple critical considerations spanning from conceptual design to clinical implementation:

Target Epitope Selection:

  • The epitope should be accessible in vivo and functionally relevant

  • For inflammatory conditions, epitopes involved in LPS binding are particularly valuable

  • The selected epitope should be conserved across relevant species if preclinical animal testing is planned

Antibody Format Optimization:

  • Humanized or fully human antibodies are preferred over chimeric antibodies to minimize immunogenicity

  • Consider the antibody isotype carefully as it affects half-life and effector functions

  • Fc engineering may be necessary to enhance or diminish specific effector functions based on therapeutic goals

Mechanism of Action Characterization:

  • Determine if the therapeutic goal requires simple blocking of CD14 function or active clearance of CD14-expressing cells

  • Evaluate if the antibody blocks LPS-CD14 interaction without triggering inflammatory responses

  • Assess if the antibody affects only membrane-bound CD14 or also soluble CD14, as this impacts therapeutic scope

Safety Profile Assessment:

  • Monitor for immunosuppressive effects, as CD14 blockade may impair innate immune responses

  • Evaluate the impact on bacterial clearance capacity

  • Assess potential for anti-drug antibody formation

  • Determine cytokine release potential, particularly important for anti-immune cell targets

Dosing and Administration Strategy:

  • Consider the timing of administration relative to disease onset (e.g., early intervention in sepsis or COVID-19)

  • Determine optimal dosing based on receptor occupancy studies

  • Evaluate single-dose versus multiple-dose regimens

The NIH trial of the anti-CD14 antibody IC14 for COVID-19 provides an exemplar of therapeutic development. The trial focuses on patients hospitalized with respiratory disease and low blood oxygen due to SARS-CoV-2 infection. The therapeutic hypothesis is that by blocking CD14 during early stages of COVID-19 respiratory disease, the antibody could temper harmful inflammatory responses, thereby limiting tissue damage and improving patient outcomes .

How can researchers evaluate the functional blockade efficacy of anti-CD14 antibodies?

Evaluating the functional blockade efficacy of anti-CD14 antibodies requires rigorous methodological approaches that assess the antibody's ability to interfere with CD14's biological functions:

LPS Binding Inhibition Assay:

  • Prepare CD14-expressing cells (primary monocytes or cell lines like THP-1)

  • Pre-incubate cells with various concentrations of the anti-CD14 antibody

  • Add fluorescently-labeled LPS (FITC-LPS)

  • Measure reduced LPS binding using flow cytometry

  • Calculate inhibition percentage using the formula:
    % Inhibition = (1 - [MFI with antibody / MFI without antibody]) × 100

Cytokine Response Inhibition:

  • Culture whole blood or isolated monocytes

  • Pre-treat with anti-CD14 antibody at varying concentrations

  • Stimulate with LPS for 4-24 hours

  • Measure pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) in culture supernatants using ELISA

  • Determine IC50 values for cytokine inhibition

Cell Signaling Pathway Analysis:

  • Pre-treat CD14-expressing cells with anti-CD14 antibody

  • Stimulate with LPS for appropriate time intervals (5-60 minutes)

  • Prepare cell lysates and analyze by Western blot

  • Measure phosphorylation of key signaling molecules (p38 MAPK, ERK, NF-κB)

  • Quantify reduction in phosphorylation relative to controls

In vitro Disease Models:

  • Establish relevant in vitro disease models (e.g., LPS-induced endothelial damage)

  • Apply anti-CD14 antibody treatment at various concentrations

  • Measure functional outcomes (e.g., endothelial permeability, tissue factor expression)

  • Compare with positive controls (known CD14 blockers) and negative controls

Receptor Occupancy Analysis:

  • Incubate cells with unlabeled test antibody at various concentrations

  • Add a different labeled anti-CD14 antibody that binds to a non-competing epitope

  • Measure by flow cytometry the reduction in binding of the labeled antibody

  • Calculate % receptor occupancy vs. antibody concentration

  • Determine EC50 for receptor occupancy

The chimeric antibody Hm2F9 provides an excellent example of functional blockade efficacy, demonstrating the ability to block LPS binding to CD14 with high efficiency, reducing positive cell percentage from 98.37% to 1.35% (a 98.63% reduction) . This level of functional blockade suggests strong potential for inhibiting CD14-mediated inflammatory responses in biological systems.

What are the optimal methods for producing monoclonal antibodies against CD14?

Production of high-quality monoclonal antibodies against CD14 involves a systematic approach from immunization to purification and validation:

Immunogen Selection and Preparation:

  • Synthetic peptide approach: Select CD14 peptides representing immunogenic epitopes, preferably from conserved regions of CD14

  • Conjugate selected peptides to carrier proteins (e.g., KLH - Keyhole Limpet Hemocyanin) to enhance immunogenicity

  • Verify conjugation efficiency using spectrophotometric methods

Immunization Protocol:

  • Use 6-8 week old female mice (BALB/c preferred)

  • Primary immunization: Emulsify 50-100 μg conjugated peptide with complete Freund's adjuvant

  • Booster immunizations: At 2-week intervals with incomplete Freund's adjuvant

  • Verify antibody titer in serum samples by ELISA before proceeding to fusion

Hybridoma Production:

  • Harvest spleen cells from immunized mice showing high antibody titers

  • Fuse spleen cells with myeloma cells (e.g., SP2/0) using polyethylene glycol

  • Culture fused cells in selective medium (HAT medium) to eliminate unfused myeloma cells

  • Screen hybrid cell supernatants for anti-CD14 antibody production by ELISA

  • Clone positive wells by limiting dilution to ensure monoclonality

Selection and Expansion:

  • Select hybridomas based on specificity, affinity, and isotype

  • Expand selected clones in culture

  • Cryopreserve early passages to maintain stable cell lines

Antibody Purification:

  • Collect hybridoma supernatant or grow selected clones in bioreactors

  • Purify antibodies using appropriate chromatography methods:

    • Protein A/G affinity chromatography for IgG isotypes

    • Size exclusion chromatography for final polishing

  • Verify purity by SDS-PAGE

Characterization:

  • Determine isotype and light chain type (e.g., IgG2a with Kappa light chain)

  • Confirm specificity by immunoblotting and flow cytometry

  • Test functional properties in relevant bioassays

Following this methodology, researchers have successfully produced well-characterized anti-CD14 monoclonal antibodies. For example, researchers produced an IgG2a antibody with Kappa light chain that demonstrated specific reactivity with CD14 in both flow cytometry and Western blot applications .

How can researchers optimize CD14 antibody validation for flow cytometry applications?

Optimizing CD14 antibody validation for flow cytometry requires a systematic approach to ensure specificity, sensitivity, and reproducibility:

Initial Titration and Optimization:

  • Perform antibody titration experiments using serial dilutions (typically 0.1-10 μg/mL)

  • Test on positive control cells (e.g., peripheral blood monocytes or THP-1 cells)

  • Determine optimal concentration by plotting signal-to-noise ratio

  • Optimize incubation conditions (time, temperature, buffer composition)

Specificity Verification:

  • Use multiple cell populations with known CD14 expression patterns:

    • High expression: Monocytes

    • Low/negative expression: Lymphocytes

    • Negative control: Cell lines like NALM-6

  • Compare staining patterns with previously validated anti-CD14 antibodies

  • Perform blocking experiments with unlabeled antibody or recombinant CD14 protein

  • Analyze with appropriate gating strategies to distinguish positive and negative populations

Cross-Reactivity Assessment:

  • Test on cells from different species if cross-reactivity is claimed

  • Verify lack of binding to similar receptors (e.g., TLR4, which associates with CD14)

  • Perform epitope mapping if necessary to confirm binding site specificity

Functional Validation:

  • Demonstrate that antibody binding correlates with known functional properties

  • Test if cells sorted based on antibody binding show expected LPS responsiveness

  • Verify if antibody binding affects cell function (neutralizing vs. non-neutralizing)

Reproducibility Testing:

  • Test inter-assay and intra-assay variability

  • Establish stable control samples for longitudinal monitoring

  • Create standardized protocols including:

    • Sample preparation methods

    • Instrument settings and compensation protocols

    • Gating strategies and analysis parameters

Application-Specific Validation:

  • For cell sorting applications: Verify post-sort purity and cell viability

  • For multi-color panels: Test for spectral overlap and optimize compensation

  • For quantitative applications: Establish calibration with known standards

Example validation results from research:
The chimeric antibody Hm2F9 demonstrated excellent specificity in flow cytometry, with 99.07% positive staining of CD14-expressing cells, comparable to its parental murine antibody 2F9 (98.86% positive cells) . This level of performance validation ensures reliable results in subsequent experimental applications.

What strategies can improve the design of anti-CD14 antibodies for increased specificity and reduced cross-reactivity?

Designing anti-CD14 antibodies with enhanced specificity and minimal cross-reactivity requires sophisticated strategies spanning epitope selection, structural engineering, and validation methodologies:

Epitope-Focused Design:

  • Conduct comprehensive sequence alignment analysis of CD14 across species to identify:

    • Highly conserved regions for broad cross-species reactivity

    • Species-specific regions for species-restricted antibodies

  • Perform structural analysis of CD14 to identify surface-exposed regions

  • Select epitopes distant from structurally similar proteins to minimize cross-reactivity

  • Prioritize epitopes involved in functional activities (e.g., LPS binding) for functional antibodies

Antibody Engineering Approaches:

  • Employ phage display technology to select high-affinity antibody variants:

    • Create diverse antibody libraries

    • Perform stringent selection against specific CD14 epitopes

    • Introduce negative selection steps against similar proteins

  • Implement affinity maturation techniques:

    • CDR (Complementarity Determining Region) walking

    • Site-directed mutagenesis of key residues

    • Computational structure-guided design

  • Consider framework modifications to reduce potential off-target interactions

Structural Refinement Strategies:

  • Utilize X-ray crystallography or cryo-EM to determine antibody-antigen complex structures

  • Apply molecular dynamics simulations to analyze binding interface stability

  • Engineer key residues at the binding interface to enhance selectivity

  • Consider smaller antibody formats (Fab, scFv) for improved epitope access in certain applications

Validation for Specificity:

  • Implement rigorous cross-reactivity testing panel:

    • Test against closely related proteins in the same family

    • Examine binding to different cell types with varying CD14 expression

    • Evaluate reactivity across different species if relevant

  • Perform competitive binding assays with known ligands

  • Utilize CD14 knockout or knockdown models as negative controls

  • Apply advanced techniques like surface plasmon resonance to quantify binding kinetics and specificity

Production Considerations:

  • Select expression systems that ensure proper folding and post-translational modifications:

    • CHO cells are preferred for full antibodies

    • Bacterial systems may be sufficient for certain antibody fragments

  • Implement stringent purification protocols to remove variants with altered specificity

  • Develop stability-indicating assays to monitor potential changes during storage

By implementing these strategies, researchers have successfully developed highly specific anti-CD14 antibodies, as demonstrated by the chimeric antibody Hm2F9, which maintained strong specific binding to CD14 while effectively blocking LPS-CD14 interactions .

How can researchers utilize anti-CD14 antibodies to study inflammatory diseases?

Anti-CD14 antibodies offer powerful tools to investigate inflammatory disease mechanisms, progression, and potential therapeutic interventions. The following methodological approaches outline how researchers can effectively utilize these antibodies in inflammatory disease research:

In Vitro Inflammatory Models:

  • LPS-induced inflammation model:

    • Isolate primary monocytes or use monocytic cell lines (THP-1)

    • Pre-treat with anti-CD14 antibodies at various concentrations

    • Challenge with LPS and measure:

      • Pro-inflammatory cytokine production (TNF-α, IL-1β, IL-6)

      • NF-κB pathway activation

      • ROS production

    • Compare effects with isotype control antibodies

  • Co-culture systems to study cell-cell interactions:

    • Establish co-cultures of monocytes with endothelial cells or epithelial cells

    • Apply anti-CD14 antibodies to block specific interactions

    • Measure adhesion, migration, and inflammatory activation markers

    • Analyze the role of CD14 in cellular crosstalk during inflammation

Ex Vivo Tissue Models:

  • Whole blood stimulation assays:

    • Collect fresh human whole blood

    • Pre-treat with anti-CD14 antibodies

    • Stimulate with various TLR ligands (LPS, peptidoglycan)

    • Analyze cytokine responses and leukocyte activation

    • Determine the contribution of CD14 to different pathogen-associated molecular pattern responses

  • Precision-cut tissue slice models:

    • Prepare tissue slices from relevant organs (lung, liver)

    • Treat with anti-CD14 antibodies prior to inflammatory stimuli

    • Assess tissue-specific inflammatory responses and damage markers

    • Evaluate CD14 role in organ-specific inflammation

In Vivo Disease Models:

  • Endotoxemia and sepsis models:

    • Administer anti-CD14 antibodies at various timepoints relative to LPS challenge

    • Monitor survival, clinical parameters, and organ dysfunction

    • Measure systemic and local cytokine responses

    • Assess vascular integrity and coagulation parameters

  • Acute lung injury/ARDS models:

    • Deliver anti-CD14 antibodies systemically or via intratracheal route

    • Challenge with inflammatory stimuli (LPS, bacteria, viral mimetics)

    • Evaluate lung function, inflammatory cell infiltration, and alveolar damage

    • Analyze bronchoalveolar lavage fluid for inflammatory mediators

  • Sterile inflammation models:

    • Induce sterile inflammation (trauma, ischemia-reperfusion)

    • Treat with anti-CD14 antibodies to assess the role of CD14 in DAMP recognition

    • Measure inflammatory parameters and tissue damage markers

    • Determine if CD14 blockade differentially affects PAMP vs. DAMP responses

Product Science Overview

Functional Role of CD14

CD14 is a pattern recognition receptor that plays a pivotal role in the innate immune system. It recognizes and binds to LPS, a component of the outer membrane of Gram-negative bacteria, facilitating the activation of immune responses . CD14 is also involved in the recognition of other microbial components, making it a critical player in the body’s defense against infections .

Applications in Research

The Mouse Anti-Human CD14 antibody is extensively used in research to study monocyte and macrophage biology, immune responses, and the mechanisms of bacterial infections . It is also used in the development of diagnostic and therapeutic strategies for various diseases involving the immune system .

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 2024 Thebiotek. All Rights Reserved.