CD14 Human

CD14 Human Recombinant
Shipped with Ice Packs
In Stock

Description

CD14 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 353 amino acids (20-349 a.a) and having a molecular mass of 37.9kDa.
CD14is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
CD14, also known as lipopolysaccharide (LPS) receptor, is primarily found on monocytes and macrophages, with weaker expression on neutrophils. It is a glycosylphosphatidylinositol (GPI)-anchored protein that serves as a high-affinity receptor for complexes of LPS and LPS binding protein (LBP). Soluble CD14 can also bind LPS, acting as an agonist at physiological concentrations and an antagonist at higher concentrations in cell activation. Additionally, CD14 has been observed to bind apoptotic cells.
Description
Recombinant human CD14, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 353 amino acids (residues 20-349). It has a molecular weight of 37.9 kDa. The protein includes a 23-amino acid His-tag at the N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that is sterile-filtered.
Formulation
The CD14 protein solution has a concentration of 0.5 mg/ml and is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 30% glycerol, 0.15 M NaCl, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Monocyte differentiation antigen CD14, Myeloid cell-specific leucine-rich glycoprotein, CD14.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSTTPEPCE LDDEDFRCVC NFSEPQPDWS EAFQCVSAVE VEIHAGGLNL EPFLKRVDAD ADPRQYADTV KALRVRRLTV GAAQVPAQLL VGALRVLAYS RLKELTLEDL KITGTMPPLP LEATGLALSS LRLRNVSWAT GRSWLAELQQ WLKPGLKVLS IAQAHSPAFS CEQVRAFPAL TSLDLSDNPG LGERGLMAAL CPHKFPAIQN LALRNTGMET PTGVCAALAA AGVQPHSLDL SHNSLRATVN PSAPRCMWSS ALNSLNLSFA GLEQVPKGLP AKLRVLDLSC NRLNRAPQPD ELPEVDNLTL DGNPFLVPGT ALPHEGSMNS GVV.

Q&A

What is the structural characterization of human CD14?

Human CD14 exhibits a bent solenoid structure typical of leucine-rich repeat proteins, with an amino-terminal pocket that binds acylated ligands including LPS. The crystal structure reveals that while human and mouse CD14 are highly superimposable (root mean square deviation: 1.089 Å), there are notable differences in charged residues on the rim and between the shape of the N-terminal pocket . For instance, the positively charged K38 residue in murine CD14 is substituted by a negatively charged D44 residue in human CD14 . The human CD14 structure was determined via X-ray crystallography at 4.0 Å resolution in the P 32 2 1 space group, and has been deposited in the Protein Data Bank (PDB ID 4GLP) . Despite these structural differences, both human and mouse CD14 likely bind similar ligands and activate concordant cellular pathways .

How does CD14 function in innate immune signaling?

CD14 functions primarily as a co-receptor for toll-like receptors (TLRs), especially TLR4, to activate innate immune responses. It recognizes and binds to pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) from gram-negative bacteria, lipoteichoic acid (LTA), and biglycan . After binding these ligands, CD14 transfers them to the TLR-MD-2 complex, promoting TLR engagement with adapter proteins like MyD88, Mal (TIRAP), TRIF, and TRAM to initiate intracellular signaling . This signaling cascade activates transcription factors such as NF-κB and IRFs in dendritic cells, ultimately leading to pro-inflammatory cytokine production . Additionally, CD14 plays a critical role in TLR4 trafficking to endosomes, which is necessary for the TRAM-TRIF-dependent pathway activation . The dual functionality of CD14 in both ligand transfer and receptor trafficking highlights its essential role as more than just a passive binding protein in innate immunity.

Which cell types express CD14 in humans?

CD14 expression occurs across a diverse range of cell types in humans. It is most abundantly expressed on cells of myeloid lineage, particularly monocytes and macrophages, where it was initially identified . Substantial expression has also been documented in neutrophils and dendritic cells, with lower levels detected in T-cells, B-cells, and liver resident Kupffer cells . Beyond the immune compartment, CD14 expression extends to several non-immune cell types including human enterocytes, hepatocytes, and pancreatic islet beta cells . This broad expression pattern across both immune and non-immune cells suggests that CD14 functions extend well beyond its canonical role in pathogen recognition, potentially participating in tissue homeostasis, metabolism, and other physiological processes that researchers are just beginning to characterize .

How should experiments be designed to investigate the differential roles of membrane-bound versus soluble CD14?

Experimental designs to distinguish between membrane-bound and soluble CD14 functions require careful methodological considerations. For in vitro studies, researchers should conduct parallel experiments using recombinant soluble CD14 versus cell lines expressing only membrane-bound CD14 (by removing the secretion signal sequence). When stimulating with LPS, measure both early responses (NF-κB activation, cytokine mRNA) and late responses (cytokine secretion, receptor internalization) to capture potential temporal differences in signaling dynamics .

For in vivo approaches, develop transgenic mouse models expressing either exclusively membrane-bound CD14 (by mutating cleavage sites) or soluble CD14 (by removing the GPI anchor sequence), then challenge with sepsis models such as cecal ligation and puncture . Monitor bacterial clearance, tissue damage markers, and survival outcomes to determine differential protective effects . When analyzing clinical samples, measure both forms simultaneously using flow cytometry for membrane-bound CD14 and ELISA for soluble CD14, correlating their ratio rather than absolute values with disease outcomes .

During experimental analysis, consider that P2X7 receptor activation induces CD14 release in extracellular vesicles, resulting in decreased cell surface CD14 that functionally affects responses to LPS but not to monophosphoryl lipid A (MPLA) . This selective effect provides a useful experimental tool to distinguish CD14-dependent from CD14-independent signaling pathways.

What experimental approaches are necessary to identify novel non-immune functions of CD14?

Identifying novel non-immune functions of CD14 requires multidisciplinary experimental approaches. Begin with transcriptomic analyses comparing CD14-expressing non-immune cells (such as enterocytes, hepatocytes, and pancreatic beta cells) with CD14-knockdown counterparts to identify differentially regulated non-immune pathways . Follow with proteomic interaction studies using proximity labeling techniques to discover CD14 binding partners unique to non-immune cells.

Functional studies should employ tissue-specific conditional CD14 knockout models that preserve immune CD14 functions while selectively deleting CD14 in tissues of interest . Phenotype these models under both homeostatic and stress conditions, focusing on non-inflammatory readouts such as metabolic parameters, barrier function, or tissue repair. For metabolic investigations, particularly relevant given CD14's emerging role in insulin sensitivity, perform glucose tolerance tests, hyperinsulinemic-euglycemic clamps, and adipose tissue analysis in mice with hepatocyte or adipocyte-specific CD14 deletion .

Ex vivo studies using primary human non-immune cells treated with soluble CD14 can help validate findings across species. Finally, correlate tissue-specific CD14 expression patterns with non-immune pathologies in patient samples, looking beyond traditional inflammatory markers to include metabolic parameters, tissue architecture, and cell-specific functional markers .

How can researchers design experiments to elucidate the mechanisms of P2X7-mediated CD14 release?

Designing experiments to elucidate P2X7-mediated CD14 release mechanisms requires a systematic approach. First, establish the temporal relationship between P2X7 activation and CD14 release through time-course experiments with ATP stimulation (2-5 mM) in wild-type versus P2rx7−/− macrophages, measuring membrane CD14 by flow cytometry and soluble CD14 by ELISA . Include P2X7 antagonists (A438079) to confirm receptor specificity.

To characterize released CD14, isolate extracellular vesicles through differential ultracentrifugation and analyze their size distribution, quantity, and CD14 content using nanoparticle tracking analysis and western blotting . Apply proteomics to identify other cargo proteins that may functionally interact with CD14 in these vesicles. Investigate the molecular machinery connecting P2X7 activation to CD14 release using pharmacological inhibitors and genetic approaches targeting vesicle formation pathways, cytoskeletal components, and membrane remodeling proteins.

For functional studies, examine how P2X7-mediated CD14 depletion affects subsequent responses to LPS versus MPLA stimulation, monitoring cytokine production (IL-6, TNF-α) at both mRNA and protein levels . Include CD14 blocking antibodies as positive controls for CD14 dependency. In vivo, compare wild-type and P2rx7−/− mice using sepsis models, measuring circulating CD14 levels, bacterial load, organ damage, and survival to establish the physiological significance of this pathway in infection responses .

What are the optimal methods for measuring and differentiating between membrane-bound and soluble CD14 in clinical samples?

Optimal measurement of both CD14 forms in clinical samples requires complementary methodological approaches. For membrane-bound CD14, flow cytometry remains the gold standard, using fluorescently-labeled antibodies against epitopes distant from the cleavage region. Sample preparation is critical—use fresh whole blood or isolated cells processed within 4 hours of collection, as delayed processing can alter surface expression levels . Include isotype controls and quantitative beads to standardize measurements across different instruments and studies.

For soluble CD14, enzyme-linked immunosorbent assay (ELISA) offers high sensitivity and specificity . When selecting commercial kits, verify capture antibody specificity against various forms of soluble CD14 (full-length versus proteolytically cleaved). For samples from septic patients, consider potential interference from lipopolysaccharide binding protein or acute phase proteins . Process plasma samples consistently, as freeze-thaw cycles can affect measurements.

To characterize vesicle-associated CD14, implement differential ultracentrifugation followed by western blotting or specialized ELISAs that selectively measure vesicle-bound versus free soluble CD14 . For comprehensive analysis, combine these approaches with multiplexed cytokine panels to contextualize CD14 measurements within the broader inflammatory environment. When monitoring therapeutic interventions, prioritize longitudinal measurements over single time points to capture dynamic changes in the membrane-bound to soluble CD14 ratio .

What techniques provide the most reliable structural analysis of human CD14?

Reliable structural analysis of human CD14 requires a multi-technique approach. X-ray crystallography has successfully resolved the human CD14 structure at 4.0 Å resolution, revealing its bent solenoid conformation with an N-terminal binding pocket for LPS . For crystallization, express recombinant human CD14 excluding the GPI-anchor region in mammalian cells to ensure proper glycosylation, followed by affinity purification using Fc-tag systems and size exclusion chromatography . Screening various crystallization conditions is essential, with successful crystals previously obtained in the P 32 2 1 space group .

For analyzing ligand interactions and conformational changes, complement crystallography with solution techniques like hydrogen-deuterium exchange mass spectrometry and small-angle X-ray scattering, which capture dynamic structural features. Circular dichroism spectroscopy provides valuable information on secondary structure content and folding stability under varying conditions.

Computational approaches including molecular dynamics simulations can model CD14 interactions with diverse ligands, particularly valuable given the challenges in crystallizing CD14-ligand complexes. For glycosylation analysis, employ mass spectrometry-based glycoproteomics to characterize site-specific glycan structures that may influence ligand binding and receptor interactions . When comparing structural data between species, consider the specific differences in charged residues and pocket architecture, such as the substitution of a positively charged K38 in mouse CD14 with a negatively charged D44 in human CD14 .

What are the most effective experimental controls when studying CD14-dependent signaling pathways?

Effective experimental controls for CD14-dependent signaling studies must address multiple potential confounding factors. First, implement genetic controls using CD14 knockout cells/animals alongside wild-type counterparts, complemented by CD14 reconstitution experiments to confirm phenotype rescue . For partial inhibition, include anti-CD14 blocking antibodies (such as clone M14-23) at validated concentrations (typically 20 μg/mL) .

Ligand-specific controls are critical—compare responses to CD14-dependent ligands (like LPS) versus CD14-independent ligands (such as MPLA) under identical experimental conditions . This comparison allows discrimination between CD14-specific effects and general alterations in cell responsiveness. When studying soluble CD14, include recombinant CD14 at physiologically relevant concentrations (0.5-5 μg/mL) alongside controls for contaminants, particularly endotoxin which can directly trigger the pathways being studied.

For cell-type specificity, use multiple cell lines/primary cells with varying CD14 expression levels, and consider species differences—human neutrophils express more CD14 than mouse neutrophils . Time-course experiments are essential as CD14 dependency may vary at different stages of the response. Finally, include pathway-specific controls targeting downstream components (TLR4, MyD88, TRIF) to distinguish direct CD14 effects from secondary consequences . When studying P2X7-mediated CD14 release, always include the specific P2X7 antagonist A438079 to confirm receptor specificity .

How should researchers analyze CD14 polymorphisms in relation to disease susceptibility?

Analysis of CD14 polymorphisms in relation to disease requires robust methodological approaches to avoid conflicting interpretations. Begin with precise genotyping methods, preferably direct sequencing or high-resolution melting analysis rather than restriction fragment length polymorphism, to accurately identify variants like the -159 C/T (rs2569190) promoter polymorphism that affects CD14 expression levels .

Statistical analysis should implement multivariate models adjusting for demographic factors, comorbidities, and environmental exposures that might influence CD14 expression independent of genetics. Use Bonferroni or false discovery rate corrections for multiple testing, and calculate appropriate sample sizes based on expected effect sizes to avoid underpowered studies.

For functional validation, measure both soluble CD14 levels and membrane-bound CD14 expression in genotyped individuals, as polymorphisms may differentially affect these forms . Conduct in vitro reporter assays with constructs containing different polymorphic variants to directly assess transcriptional impacts. Importantly, perform stratified analyses based on disease etiology, as the same CD14 variant may have opposing effects in different inflammatory contexts .

Meta-analytical approaches incorporating rigorous study quality assessment are essential when interpreting apparently contradictory results across populations. Finally, consider gene-environment and gene-gene interactions, particularly with TLR4 polymorphisms, as these may explain population-specific associations between CD14 variants and disease susceptibility .

What approaches help resolve contradictory findings on CD14's role in infectious versus sterile inflammation?

Resolving contradictory findings on CD14's role across different inflammatory contexts requires systematic analytical approaches. First, standardize experimental models and readouts when comparing infectious versus sterile inflammation, using the same genetic backgrounds, time points, and analytical methods to ensure valid comparisons . Conduct parallel studies with both types of inflammation in the same experimental system to control for laboratory-specific variables.

Distinguish between early and late phases of inflammatory responses, as CD14 may have phase-specific roles—initially promoting pathogen recognition while later regulating resolution processes . Apply systems biology approaches integrating transcriptomics, proteomics, and metabolomics to map context-specific CD14 signaling networks in different inflammatory scenarios .

When analyzing clinical data, stratify patients by inflammation etiology (infectious, autoimmune, metabolic) and perform subgroup analyses to identify context-specific associations . Consider the balance between membrane-bound and soluble CD14, as their ratio rather than absolute levels may determine outcomes . Specifically examine how P2X7 receptor-mediated CD14 release affects this balance in different inflammatory settings .

For mechanistic resolution, investigate ligand-specific effects—different PAMPs and DAMPs may engage CD14 differently despite converging on similar downstream pathways. Finally, consider the tissue microenvironment, as CD14 functions may differ substantially between compartments (circulation versus tissue-resident) and cell types (myeloid versus epithelial) .

How can researchers distinguish between correlation and causation when analyzing CD14 levels in disease states?

Establishing causality beyond correlation for CD14 in disease states requires methodological rigor. Implement temporal analysis through longitudinal studies measuring CD14 levels before disease onset and during progression to establish whether CD14 changes precede clinical manifestations . Use Mendelian randomization approaches with CD14 genetic variants as instrumental variables to mitigate confounding factors when analyzing observational data.

Experimental manipulation through intervention studies provides stronger evidence—CD14 blockade with antibodies or genetic deletion should alter disease trajectory if CD14's relationship is causal . Apply dose-response testing with recombinant CD14 in cellular and animal models to establish biological gradients consistent with causality.

Mechanistic validation is essential—proposed causal pathways should be systematically tested through inhibitor studies and genetic approaches targeting specific steps in the pathway . Use mediation analysis to quantify how much of CD14's effect on outcomes occurs through specific mechanisms versus alternative pathways.

Cross-validation across multiple model systems strengthens causal inference—findings should be consistent across in vitro systems, animal models, and human studies . When contradictions arise, investigate context-dependent factors that might explain differential effects. Finally, implement experimental designs specifically addressing alternative explanations for observed associations, such as reverse causality (where disease causes CD14 changes) or confounding by inflammatory status, using appropriate statistical adjustments and experimental controls .

What approaches should researchers take to develop CD14-targeted therapeutics for sepsis?

Developing CD14-targeted therapeutics for sepsis requires nuanced approaches recognizing CD14's complex roles. Begin with comprehensive target validation, determining whether to inhibit membrane-bound CD14, neutralize soluble CD14, or modulate the balance between forms . Evidence suggests complete CD14 blockade may be detrimental, as P2X7-mediated CD14 release maintains elevated circulating CD14 during infection, which appears necessary for bacterial clearance .

For antibody development, epitope mapping is critical—different CD14 regions mediate distinct functions, offering the potential for selective inhibition of pathological rather than protective activities . Consider bispecific antibodies targeting both CD14 and damage-associated molecular patterns that emerge during sepsis to specifically inhibit sterile inflammation components.

Pharmacological timing is crucial—sepsis progresses through distinct phases with potentially different optimal intervention points for CD14 targeting . Design adaptive dosing strategies based on real-time monitoring of soluble CD14 levels and inflammatory markers. For preclinical evaluation, utilize multiple sepsis models beyond endotoxemia, including polymicrobial sepsis through cecal ligation and puncture, as these better recapitulate the clinical complexity .

Patient stratification strategies should incorporate both CD14 genetic polymorphisms and baseline soluble CD14 levels, as these may predict response to CD14-targeted interventions . Consider complementary approaches targeting P2X7 receptors to modulate CD14 release during sepsis, as research indicates P2X7 is important for maintaining elevated circulating CD14 during infection, helping to clear bacteria and prevent excessive organ damage .

How can CD14 be effectively investigated as a biomarker for inflammatory diseases?

Investigating CD14 as an inflammatory biomarker requires methodological precision to establish clinical utility. First, standardize measurement protocols across laboratories using reference materials and validated assays that distinguish between different forms of CD14 (membrane-bound, soluble, vesicle-associated) . Establish reference ranges in healthy populations stratified by age, sex, and ethnicity, accounting for common CD14 polymorphisms that affect baseline levels .

Conduct longitudinal studies in at-risk populations to determine whether CD14 changes precede clinical manifestations, establishing its prognostic value. Calculate sensitivity, specificity, and predictive values compared to existing biomarkers, and combine CD14 with other markers in multiparametric panels to improve diagnostic accuracy . Investigate whether the ratio of different CD14 forms provides superior diagnostic information compared to absolute levels of any single form .

For specific diseases, validate CD14 in independent cohorts with diverse characteristics and disease severities. In sepsis research, determine whether soluble CD14 dynamics correlate with bacterial clearance and organ protection, consistent with its proposed role in mediating P2X7-dependent protective effects . For metabolic diseases, investigate relationships between CD14 levels, insulin sensitivity, and adipose tissue inflammation .

Standardize reporting using STARD (Standards for Reporting of Diagnostic Accuracy) guidelines to facilitate meta-analyses and systematic reviews. Finally, conduct interventional studies where treatment decisions are based on CD14 levels to determine whether biomarker-guided approaches improve clinical outcomes compared to standard care .

What are the key translational challenges in applying mouse CD14 findings to human disease?

Expression pattern differences must be considered—human neutrophils express higher CD14 levels than their murine counterparts, and tissue-specific expression patterns vary between species . When developing therapeutics, these differences necessitate careful epitope selection and validation in human systems before clinical translation.

Inflammatory responses show species-specific features—human and mouse macrophages differ in cytokine profiles and metabolic responses to CD14-dependent stimuli . LPS sensitivity varies substantially between species, with humans showing greater sensitivity than mice, potentially due to differences in CD14-TLR4-MD2 complex interactions .

For disease modeling, recognize that mouse models often represent acute conditions, while human diseases like sepsis involve complex comorbidities and chronic inflammatory states . Humanized mouse models expressing human CD14 can bridge some translational gaps but have limitations in recapitulating the human immune environment.

Address these challenges by implementing parallel human in vitro systems alongside mouse models, validating key findings in primary human cells and tissues . When possible, confirm mechanistic insights from mouse models using patient samples, particularly for processes like P2X7-mediated CD14 release, which may have species-specific regulation .

How can researchers investigate CD14's expanding roles in metabolic regulation?

Investigating CD14's metabolic functions requires targeted methodological approaches beyond traditional immunological techniques. Develop tissue-specific CD14 knockout models, particularly targeting adipose tissue, liver, and pancreatic β-cells, where CD14 expression has been detected in humans . Perform comprehensive metabolic phenotyping including glucose and insulin tolerance tests, hyperinsulinemic-euglycemic clamps, and metabolic cage analyses to assess energy expenditure and substrate utilization.

At the cellular level, use stable isotope tracing to map how CD14 signaling influences metabolic flux through glycolysis, fatty acid oxidation, and gluconeogenesis pathways. Investigate how CD14 affects adipose insulin sensitivity and lipid storage by measuring insulin-stimulated glucose uptake and lipolysis in primary adipocytes with CD14 deletion or overexpression .

For translational relevance, analyze relationships between CD14 genetic variants, circulating CD14 levels, and metabolic parameters in human cohorts with detailed metabolic phenotyping . Investigate how weight loss interventions affect CD14 expression and signaling in metabolic tissues. Explore CD14's potential role in diet-induced metabolic inflammation by examining how different dietary components affect CD14-dependent signaling in hepatocytes and adipocytes.

Finally, investigate cross-talk between CD14 and key metabolic hormones (insulin, leptin, adiponectin) using co-immunoprecipitation, proximity ligation assays, and receptor trafficking studies to uncover potential direct interactions that might explain CD14's emerging roles in adrenal function and glucose responsiveness .

What methodological approaches can best identify novel CD14 ligands beyond classical PAMPs?

Identifying novel CD14 ligands requires systematic screening approaches beyond traditional candidate testing. Implement unbiased biochemical screens using recombinant human CD14 immobilized on biosensor chips for surface plasmon resonance analysis, allowing real-time detection of binding interactions with complex biological samples from different disease states . Follow with pull-down assays and mass spectrometry to identify specific binding partners.

Leverage the crystal structure of human CD14 for in silico screening and molecular docking approaches, focusing on the N-terminal pocket and considering the specific features of human CD14 that differ from the mouse ortholog, such as the negatively charged D44 residue . This computational approach can prioritize candidates for experimental validation.

For damage-associated molecular patterns (DAMPs), which may represent an important class of endogenous CD14 ligands, develop cell stress models that generate defined damage signals and test their binding to CD14 using competition assays with known ligands like LPS . Investigate whether oxidation or other modifications alter the ability of endogenous molecules to bind CD14, potentially explaining how tissue damage triggers CD14-dependent responses.

Employ cellular screening systems using reporter cells expressing human CD14 coupled to downstream signaling readouts. Test fractionated human tissue extracts from various pathological conditions to identify activating components . Finally, develop chemical biology approaches using photoactivatable crosslinking probes based on known CD14 ligands to capture transient binding interactions that might be missed by traditional binding assays .

How can researchers investigate the therapeutic potential of modulating P2X7-mediated CD14 release?

Investigating P2X7-CD14 therapeutic potential requires methodological approaches targeting this specific release mechanism. First, characterize the molecular requirements for P2X7-mediated CD14 release using pharmacological inhibitors of various vesicle formation pathways and genetically modified cells lacking key components of the secretory machinery . This will identify specific druggable targets besides P2X7 itself.

Develop small molecules or peptides that selectively modulate P2X7-mediated CD14 release without affecting other P2X7 functions, using high-throughput screening with cell lines expressing fluorescently tagged CD14 . Validate lead compounds in primary human macrophages, monitoring both CD14 release and subsequent functional consequences for LPS responsiveness.

For in vivo proof-of-concept, test modulators in clinically relevant sepsis models, measuring bacterial clearance, organ damage markers, and survival alongside CD14 levels in circulation . Compare outcomes with direct CD14 manipulation (antibodies, recombinant protein) to determine whether targeting the release mechanism offers advantages over targeting CD14 itself.

Investigate whether impaired P2X7-mediated CD14 release contributes to human inflammatory diseases by analyzing P2X7 polymorphisms in patient cohorts alongside soluble CD14 measurements . Develop ex vivo assays using patient-derived cells to test whether P2X7 agonists can restore beneficial CD14 release patterns in those with dysregulated inflammation.

Finally, consider combination approaches targeting both P2X7-mediated CD14 release and downstream CD14 signaling at different disease stages . For example, in sepsis, enhancing CD14 release during early bacterial clearance while dampening CD14-dependent inflammation during the later cytokine storm phase could potentially address the complex, dynamic nature of inflammatory diseases .

Product Science Overview

Structure and Function

The human CD14 protein is encoded by the CD14 gene and consists of 375 amino acids . It includes a 19 amino acid signal peptide and a C-terminal hydrophobic region characteristic of glycosylphosphatidylinositol (GPI)-anchored proteins . The GPI anchor allows CD14 to be attached to the cell membrane, although a soluble form of CD14 (sCD14) also exists in the serum .

CD14 enhances the immune response by binding to LPS and facilitating its recognition by the Toll-like receptor 4 (TLR4)-MD-2 complex . This interaction triggers a cascade of signaling events leading to the production of pro-inflammatory cytokines and the activation of the immune system .

Recombinant CD14

Recombinant human CD14 is produced using various expression systems, including Chinese Hamster Ovary (CHO) cells and HEK293 cells . The recombinant protein is often tagged with a His-tag to facilitate purification and detection . It is used in research to study the immune response, particularly the mechanisms of LPS recognition and signaling.

The recombinant CD14 protein is typically purified to a high degree of purity (>95%) and is tested for endotoxin levels to ensure its suitability for experimental use . It is available in both carrier-free and carrier-containing formulations, depending on the specific application requirements .

Applications

Recombinant CD14 is widely used in immunological research to:

  • Study the interaction between LPS and the TLR4-MD-2 complex.
  • Investigate the signaling pathways activated by LPS.
  • Explore the role of CD14 in various immune responses and diseases.

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.