LBP Human Recombinant is a single, glycosylated polypeptide chain containing 462 amino acids (26-481a.a) and having a molecular mass of 51.7kDa (calculated). LBP is fused to a 6 a.a His tag at C-terminal.
Lipopolysaccharide-binding protein, LBP, MGC22233, Ly88.
HEK293 Cells.
ANPGLVARIT DKGLQYAAQE GLLALQSELL RITLPDFTGD LRIPHVGRGR YEFHSLNIHS CELLHSALRP VPGQGLSLSI SDSSIRVQGR WKVRKSFFKL QGSFDVSVKG ISISVNLLLG SESSGRPTVT ASSCSSDIAD VEVDMSGDLG WLLNLFHNQI ESKFQKVLES RICEMIQKSV SSDLQPYLQT LPVTTEIDSF ADIDYSLVEA PRATAQMLEV MFKGEIFHRN HRSPVTLLAA VMSLPEEHNK MVYFAISDYV FNTASLVYHE EGYLNFSITD DMIPPDSNIR LTTKSFRPFV PRLARLYPNM NLELQGSVPS APLLNFSPGN LSVDPYMEID AFVLLPSSSK EPVFRLSVAT NVSATLTFNT SKITGFLKPG KVKVELKESK VGLFNAELLE ALLNYYILNT FYPKFNDKLA EGFPLPLLKR VQLYDLGLQI HKDFLFLGAN VQYMRVHHHH HH.
Human Lipopolysaccharide-binding protein (LBP) is an acute-phase protein that plays a critical role in the innate immune response to bacterial infections. LBP specifically binds to the lipid A moiety of bacterial lipopolysaccharides (LPS), which is a glycolipid present in the outer membrane of all Gram-negative bacteria . This binding is essential for the initial recognition of bacterial pathogens.
Functionally, LBP acts as an affinity enhancer for CD14, facilitating its association with LPS. This interaction is crucial for downstream signaling cascades that lead to the activation of immune responses. LBP effectively catalyzes the transfer of LPS to CD14, which then presents LPS to the TLR4/MD-2 complex on cell surfaces . This process ultimately promotes the release of cytokines in response to bacterial lipopolysaccharide, orchestrating appropriate inflammatory responses to infection .
The molecular structure of LBP includes specific binding domains that allow it to recognize and interact with LPS with high specificity, making it an important mediator in the early detection of Gram-negative bacterial invasion.
HEK293 cells have become a widely used expression host for transient gene expression in biomedical research for several compelling scientific reasons. These human embryonic kidney-derived cells offer a eukaryotic expression system that provides proper post-translational modifications for human proteins .
The popularity of HEK293 cells stems from several key advantages:
High transfection efficiency compared to other mammalian cell lines
Relatively robust growth characteristics in various culture conditions
Capacity to express complex proteins with proper folding and assembly
Ability to produce high yields of recombinant proteins
Human-derived cellular machinery that ensures appropriate processing of human proteins
These characteristics make HEK293 cells particularly valuable for the expression of complex human proteins like antibodies and membrane receptors. For example, adalimumab has been successfully expressed in HEK293 cells with high yields, making it an excellent model protein for recombinant expression studies . The human cellular environment ensures that expressed proteins maintain native conformations and functional properties, which is essential for accurate biological characterization and therapeutic development.
The effect of LPS contamination on HEK293 cell cultures varies significantly depending on the concentration level. Systematic studies have revealed a dose-dependent relationship between LPS exposure and cell viability as well as protein expression outcomes .
At low concentrations (5-50 EU/mL calculated values), LPS appears to be well-tolerated by HEK293 cells and may even enhance protein expression in some cases. This unexpected finding suggests that mild LPS exposure might trigger cellular mechanisms that temporarily boost protein synthesis machinery .
At moderate concentrations (around 500 EU/mL), HEK293 cells can generally maintain normal growth patterns without major distinctive features in either transiently transfected or non-transfected cultures .
The most striking observation is that in some experimental settings, low amounts of added LPS (5–50 EU/mL calculated) can actually increase protein expression. This may occur in situations where cells are challenged with high-level recombinant protein expression and cannot cope with folding and assembly demands. The mild stress from low LPS levels might trigger cellular responses that enhance protein processing capacity .
The molecular mechanisms governing LPS effects on recombinant protein expression in HEK293 cells involve a complex interplay between cellular stress responses, innate immune signaling, and protein synthesis machinery. Though HEK293 cells are not immune cells per se, they express endogenous levels of various pattern recognition receptors (PRRs), including TLR3, TLR5, and RIG-I-like receptors .
When LPS interacts with HEK293 cells, it can trigger multiple signaling cascades:
NF-κB pathway activation: Even at low levels, LPS can activate mild NF-κB signaling, which paradoxically may upregulate certain cellular processes beneficial for protein production.
Unfolded protein response (UPR): Low LPS concentrations may induce a moderate UPR that increases the cell's capacity to handle high protein loads by upregulating chaperones and folding machinery.
Mitochondrial stress responses: LPS can alter mitochondrial function, affecting cellular energy production necessary for protein synthesis.
Translation efficiency modulation: LPS may influence translation initiation factors and ribosomal assembly, directly impacting protein synthesis rates.
At higher concentrations, these mechanisms become detrimental, shifting from adaptive responses to cytotoxic effects. The transition from beneficial to harmful effects occurs between 500 and 5000 EU/mL (calculated values), where excessive inflammatory signaling and cellular stress overwhelm adaptive responses .
The differential response observed between transfected and non-transfected cells suggests that cells already engaged in high-level protein production may have altered stress response thresholds or preactivated adaptive mechanisms that provide temporary resistance to LPS toxicity at certain concentrations.
Distinguishing LPS-induced effects from other experimental variables requires a systematic approach and appropriate controls. Researchers should implement the following methodological strategies:
Endotoxin testing: Quantify LPS concentrations in plasmid preparations using validated assays such as the Limulus Amebocyte Lysate (LAL) test. This provides actual measured values rather than calculated estimates, which is critical as measured LPS levels often differ from theoretical calculations .
Dose-response experiments: Establish a dose-response curve specific to your experimental system by testing serial dilutions of purified LPS alongside your experimental conditions.
LPS neutralization controls: Include experimental groups with LPS neutralizing agents such as polymyxin B or LPS-RS (TLR4 antagonist) to confirm observed effects are LPS-dependent.
Parallel non-transfected controls: Maintain parallel cultures of non-transfected cells exposed to identical LPS concentrations to differentiate between general cellular toxicity and transfection-specific effects .
Time-course analysis: Monitor cellular responses and protein expression at multiple time points, as LPS effects can evolve over time.
Pathway inhibition studies: Use specific inhibitors of LPS-activated pathways (e.g., NF-κB inhibitors) to determine which signaling cascades mediate observed effects.
Transcript analysis: Perform quantitative PCR to detect LPS-induced gene expression changes that may correlate with altered protein expression.
Secretome analysis: Analyze culture supernatants for inflammatory mediators that might influence cellular behavior independently of direct LPS actions.
The comparison between calculated and measured LPS levels is particularly important, as research has shown significant discrepancies between these values. For rigorous experimental design, actual measurements rather than theoretical calculations should be used to establish true exposure levels .
HEK-Blue hTLR4 cells represent a sophisticated reporter system for studying LPS-LBP interactions with high sensitivity and specificity. These engineered cells express human TLR4, along with its co-adaptors MD-2 and CD14, as well as an inducible secreted embryonic alkaline phosphatase (SEAP) reporter gene downstream of NF-κB activation .
Optimal experimental conditions for utilizing this system include:
When designing experiments, it's crucial to remember that HEK293 cells express endogenous levels of various PRRs including TLR3, TLR5, and RIG-I-like receptors, which could potentially respond to other microbial components . Therefore, highly purified LPS preparations with endotoxin levels <1 EU/μg are recommended .
For comprehensive analysis of TLR4 signaling, researchers should consider that TLR4 triggers both NF-κB and IRF pathways. While HEK-Blue hTLR4 cells report only on NF-κB activation, HEK-Dual hTLR4 cells offer dual reporters for both pathways, providing more complete mechanistic insights into LPS-LBP signaling .
Minimizing LPS contamination in plasmid DNA preparations is critical for reliable and reproducible results in HEK293 transfection experiments. The following comprehensive protocol addresses key steps for endotoxin reduction:
Bacterial strain selection:
Use endotoxin-reduced E. coli strains such as ClearColi BL21(DE3) that produce modified LPS with substantially reduced endotoxicity
Alternatively, use standard laboratory strains (DH5α, TOP10) but implement rigorous downstream purification
Growth conditions optimization:
Limit bacterial culture density (OD600 <2.0) to reduce LPS release
Harvest bacteria in early to mid-log phase rather than stationary phase
Use endotoxin-free media components and water throughout the process
Plasmid isolation methods:
Commercial endotoxin-free plasmid preparation kits that incorporate specific steps to remove LPS
Implement additional purification steps beyond standard kit protocols:
Triton X-114 phase separation (2% v/v, repeated extractions)
High-salt washes (1.5-2.0 M NaCl) to disrupt LPS-DNA interactions
Polymyxin B affinity chromatography for specific LPS binding
Quality control for LPS contamination:
Quantify endotoxin levels using LAL assay (kinetic-chromogenic or recombinant Factor C methods)
Target endotoxin levels <5 EU/mL for sensitive applications
Include positive and negative controls in LAL testing to ensure accuracy
Storage considerations:
Store purified plasmids in endotoxin-free TE buffer or water
Use endotoxin-free microcentrifuge tubes
Avoid repeated freeze-thaw cycles that may release LPS from micelles
Implementing this protocol can consistently yield plasmid preparations with endotoxin levels <1 EU/μg DNA, well below the threshold where LPS begins to affect HEK293 cell performance (5-50 EU/mL) . For particularly sensitive applications, additional steps such as cesium chloride gradients or size-exclusion chromatography may further reduce endotoxin levels.
Accurate quantification of LPS levels is essential for interpreting experimental results in HEK293 cell systems. Multiple complementary methodologies are recommended for comprehensive LPS assessment:
Limulus Amebocyte Lysate (LAL) assays:
Chromogenic endpoint: Provides quantitative results with sensitivity to 0.01-0.1 EU/mL
Kinetic turbidimetric: Offers dynamic range of 0.01-100 EU/mL with reduced protein interference
Recombinant Factor C (rFC): Eliminates false positives from fungal contamination with comparable sensitivity
HEK-Blue hTLR4 cell-based bioassay:
Utilizes engineered HEK293 cells expressing TLR4/MD-2/CD14 complex and an NF-κB-inducible SEAP reporter
Particularly valuable for determining bioactive LPS levels
Provides physiologically relevant measurements of LPS activity rather than just concentration
Detection range typically 0.01-100 ng/mL LPS
Endotoxin removal verification:
Spike-recovery tests to validate endotoxin removal procedures
Pre- and post-treatment measurements to calculate removal efficiency
Mass spectrometry approaches:
MALDI-TOF MS for structural characterization of LPS
LC-MS/MS for quantification of specific lipid A structures
When measuring LPS in complex biological samples, researchers should account for potential inhibitory or enhancing factors. For HEK293 culture media, components such as serum proteins can mask LPS detection in LAL assays. Sample dilution series, spike recovery controls, and sample treatments (heat inactivation at 70°C for 10 minutes) can help overcome these limitations.
It's important to note that calculated and measured LPS values often differ significantly. In experimental studies, calculated values of 5-50 EU/mL LPS addition may result in measured concentrations of 1-10 EU/mL due to binding of LPS to medium components or experimental surfaces . This underscores the importance of actually measuring LPS levels after addition to culture medium rather than relying solely on calculated values.
The interaction between LBP and LPS has significant implications for experimental design in HEK293-based recombinant protein production. Understanding and accounting for these interactions can improve experimental outcomes and data interpretation:
LBP as an LPS transfer catalyst:
Human LBP acts as an affinity enhancer for CD14, facilitating the transfer of LPS to CD14 and subsequent TLR4/MD-2 complex formation
This catalytic function means that even small amounts of LPS can trigger significant cellular responses if LBP is present
Researchers should measure or control for endogenous LBP in serum-containing media
LPS detection and neutralization strategies:
Recombinant human LBP (<1 EU/μg endotoxin level) can be used to study LPS-induced effects in controlled experiments
Anti-LBP neutralizing antibodies can block LPS transfer to CD14, isolating LBP-dependent effects
LBP-knockout experiments (via CRISPR/Cas9) can definitively establish LBP-dependent responses
Media composition considerations:
Serum contains variable levels of LBP that can enhance LPS sensitivity
Defined media formulations with recombinant LBP allow precise control of LPS-LBP interactions
Low-protein or protein-free formulations reduce interference with LPS-binding proteins
Experimental design guidelines for LBP-LPS studies:
Protein quality considerations:
Low LPS levels (5-50 EU/mL) may enhance protein yield but could potentially affect protein quality
Include product quality assessments (activity assays, structural analysis) alongside yield measurements
Monitor stress response indicators that might correlate with altered post-translational modifications
Research indicates that while low LPS levels might temporarily boost protein expression, they do so by triggering cellular stress responses that could affect protein quality . Therefore, comprehensive experimental designs should include both quantitative (yield) and qualitative (function, structure) assessments of recombinant proteins produced under different LPS exposure conditions.
Contradictory results regarding LPS effects on HEK293 cells are common in the literature and can be systematically analyzed through a comprehensive troubleshooting approach. Several key factors may contribute to experimental discrepancies:
LPS concentration discrepancies:
Calculated versus measured concentrations often differ significantly
Studies reporting only calculated values may be misleading as actual exposure levels can be substantially lower
Experimental setups differ considerably, making head-to-head comparison difficult
Recommendation: Always measure actual LPS concentrations in the final experimental system
LPS structural heterogeneity:
Cell line variations:
HEK293 sublines (HEK293T, HEK293F, etc.) have genetic and phenotypic differences
Passage number and culture history affect receptor expression levels
Clone-specific variations in TLR4 pathway components
Recommendation: Maintain detailed cell line provenance records
Transfection variables:
Transfection method alters cellular stress state and LPS sensitivity
LPS effects differ between transfected and non-transfected cultures
Plasmid properties (size, GC content, sequence elements) interact with LPS effects
Recommendation: Use consistent transfection protocols and include appropriate controls
Experimental timeline:
Temporal aspects of LPS exposure (acute vs. chronic)
Time of measurement relative to LPS addition
Cell density and growth phase during exposure
Recommendation: Perform time-course analyses to capture dynamic responses
When evaluating contradictory results in literature, researchers should construct a comparative analysis table:
Study | LPS Source | Concentration (Measured/Calculated) | Cell Line | Transfection Method | Exposure Duration | Key Findings |
---|---|---|---|---|---|---|
Study A | E. coli O111:B4 | 50 EU/mL (measured) | HEK293 | Polyethylenimine | 72 hours | Enhanced protein expression |
Study B | E. coli O55:B5 | 100 EU/mL (calculated) | HEK293T | Lipofectamine | 48 hours | No significant effect |
Study C | Salmonella | 5000 EU/mL (measured) | HEK293 | Calcium phosphate | 24 hours | Decreased viability |
This structured approach to analyzing experimental variables can reconcile apparently contradictory results by identifying critical differences in experimental design. The most striking finding from current research suggests that low LPS amounts (5-50 EU/mL calculated) can enhance protein expression under specific conditions, while higher concentrations are consistently detrimental .
Monitoring LPS-induced effects in HEK293 cells requires a multi-parameter approach with reliable cellular and molecular markers. These markers provide comprehensive insights into LPS responses across different biological levels:
Cellular viability and proliferation markers:
Stress response indicators:
Reactive oxygen species (ROS): Measured via DCFDA or similar fluorescent probes
Mitochondrial membrane potential: JC-1 or TMRE dyes indicate mitochondrial health
Heat shock proteins (HSP70, HSP90): Western blot or qPCR analysis of stress response
Unfolded protein response (BiP/GRP78, XBP1 splicing): Indicates ER stress levels
Signaling pathway activation:
NF-κB nuclear translocation: Immunofluorescence or nuclear fraction Western blot
SEAP reporter activity: In HEK-Blue hTLR4 cells provides quantitative NF-κB activation measurement
Phosphorylation of pathway components: p-IκB, p-p38, p-JNK via Western blot
Gene expression changes: qPCR for IL-8, TNF-α, RANTES as TLR4-responsive genes
Protein production parameters:
Total protein yield: Quantified by appropriate assays (ELISA, functional assays)
Protein quality: Size exclusion chromatography, thermal stability, activity assays
Protein folding efficiency: Ratio of properly folded to misfolded protein
Glycosylation patterns: Lectin binding assays or mass spectrometry
Integrated bioassays:
The most reliable approach combines multiple marker types to create a comprehensive profile of cellular responses. For example, research demonstrates that transiently transfected HEK293 cultures exposed to 5000 EU/mL LPS show reduced cell proliferation compared to lower LPS concentrations, yet maintain better viability than non-transfected cultures at the same LPS concentration . This finding highlights the importance of measuring multiple parameters simultaneously to capture complex cellular responses.
Researchers working with LBP and HEK293 systems should adopt a systematic approach that integrates multiple methodological considerations to ensure experimental rigor and reproducibility. The complex interplay between LPS, LBP, and HEK293 cells demands careful experimental design and interpretation.
The most critical considerations include:
LPS concentration management:
LBP-LPS interaction dynamics:
HEK293 cell line considerations:
Experimental validation approaches:
Include comprehensive controls (non-transfected cells, LPS-free conditions, LPS neutralization)
Employ multiple complementary methodologies for key measurements
Document all experimental variables that might influence outcomes
Data interpretation framework:
Consider context-specific effects based on experimental conditions
Acknowledge temporal dynamics of cellular responses to LPS
Analyze both quantitative outputs and qualitative aspects of cellular responses
Future research directions should include systematic characterization of the molecular mechanisms underlying the paradoxical enhancement of protein expression by low LPS levels, development of standardized protocols for endotoxin-free plasmid preparation, and creation of improved reporter systems for simultaneous monitoring of multiple LPS-activated pathways in HEK293 cells.
By adhering to these considerations, researchers can maximize the value of LBP-HEK293 experimental systems while minimizing confounding variables that complicate data interpretation and reproducibility.
Several emerging technologies are poised to significantly advance research on LBP-HEK interactions, opening new avenues for more precise, comprehensive, and physiologically relevant studies:
CRISPR-engineered cellular models:
CRISPR/Cas9 knockout of endogenous TLRs and adaptors in HEK293 cells
Precise insertion of reporter elements at endogenous loci
Generation of isogenic cell lines differing only in specific pathway components
Humanized receptor expression systems with physiologically relevant regulation
Advanced biosensor technologies:
Real-time, single-cell monitoring of LPS responses
FRET-based sensors for protein-protein interactions in the TLR4 pathway
Label-free detection systems for LPS-LBP-CD14-TLR4 complex formation
Microfluidic systems for controlled exposure to LPS gradients
Multi-omics approaches:
Integrated transcriptomics, proteomics, and metabolomics of LPS responses
Spatial transcriptomics for subcellular resolution of signaling responses
Temporal multi-omics to capture dynamic cellular state transitions
Systems biology modeling of LPS-induced signaling networks
Advanced structural biology methods:
Cryo-EM structures of LBP-LPS complexes at near-atomic resolution
Hydrogen-deuterium exchange mass spectrometry for dynamic binding interactions
Single-molecule FRET to track conformational changes during LPS transfer
Microphysiological systems:
Organ-on-chip technologies incorporating HEK293 expression systems
Co-culture models with immune cells to study paracrine signaling
Microfluidic systems with controlled fluid dynamics mimicking in vivo conditions
3D organoid models for studying LPS responses in tissue-like environments
These emerging technologies will enable researchers to address several outstanding questions in the field:
How does the molecular conformation of LBP change upon LPS binding?
What is the precise mechanism by which low LPS concentrations enhance protein expression in HEK293 cells?
How do post-translational modifications of LBP affect its function in LPS transfer?
What are the dynamic changes in subcellular localization of signaling components during LPS responses?
How does the cellular microenvironment modulate LBP-LPS interaction outcomes?
LBP is a glycoprotein that binds to the lipid A moiety of bacterial lipopolysaccharides (LPS), which are components of the outer membrane of Gram-negative bacteria . This binding is essential for the immune system to recognize and respond to bacterial infections. LBP acts as an affinity enhancer for CD14, facilitating its association with LPS and promoting the release of cytokines in response to bacterial lipopolysaccharide .
Recombinant Human LBP is often expressed in HEK 293 cells, a human embryonic kidney cell line. This expression system is chosen for its ability to produce high-quality, biologically active proteins. The recombinant LBP typically has a purity of over 95% and an endotoxin level of less than 1 EU/µg, making it suitable for various applications such as SDS-PAGE and HPLC .
LBP plays a pivotal role in the innate immune response by binding to LPS and transferring it to its receptor, CD14. This interaction leads to an active inflammatory response, which is crucial for fighting bacterial infections . Additionally, LBP acts as an opsonin, enhancing the phagocytosis of Gram-negative bacteria by neutrophils and granulocytes .