Lbp Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship orders within 1-3 business days of receipt. Delivery times may vary depending on the method of purchase and location. Please consult your local distributor for specific delivery information.
Synonyms
LbpLipopolysaccharide-binding protein antibody; LBP antibody
Target Names
Lbp
Uniprot No.

Target Background

Function
Lbp Antibody plays a key role in the innate immune response. It binds to the lipid A moiety of bacterial lipopolysaccharides (LPS), a glycolipid found in the outer membrane of all Gram-negative bacteria. This antibody acts as an affinity enhancer for CD14, facilitating its association with LPS. Ultimately, it promotes the release of cytokines in response to bacterial lipopolysaccharide.
Database Links
Protein Families
BPI/LBP/Plunc superfamily, BPI/LBP family
Subcellular Location
Secreted. Cytoplasmic granule membrane.
Tissue Specificity
Detected in lung, liver, heart and kidney.

Q&A

What is LBP and what is its significance in immunological research?

Lipopolysaccharide-binding protein (LBP) is a critical acute-phase protein synthesized predominantly by hepatocytes that plays a fundamental role in innate immune responses to Gram-negative bacteria. LBP functions by binding to the lipid A moiety of bacterial lipopolysaccharides (LPS), a glycolipid component in the outer membrane of all Gram-negative bacteria . In normal serum, LBP is present at concentrations of 5-10 μg/ml, but during acute-phase response, levels can rise dramatically to approximately 200 μg/ml within 24 hours . LBP's significance stems from its role as an affinity enhancer for CD14, facilitating its association with LPS and subsequently promoting cytokine release in response to bacterial endotoxins . The protein is structurally reported to be approximately 53.4 kilodaltons in mass, though observed molecular weights in laboratory contexts often range between 53-67 kDa depending on post-translational modifications and experimental conditions .

How do LBP antibodies contribute to studies of innate immunity?

LBP antibodies serve as essential tools for studying host-pathogen interactions and innate immune responses. Research has demonstrated that LBP antibodies can be strategically employed to investigate the mechanisms of endotoxin recognition and cellular activation pathways. Notably, experimental studies have revealed that different classes of anti-LBP monoclonal antibodies can target specific functional domains of LBP, providing insights into its molecular interactions . Class 1 antibodies block LPS binding to LBP, while class 2 antibodies prevent LPS/LBP complexes from interacting with CD14, and class 3 antibodies bind to LBP without suppressing its activity . In vivo studies have shown that both class 1 and class 2 antibodies suppress LPS-induced TNF production and protect mice from lethal endotoxemia, confirming LBP's critical role in innate immunity . These findings illustrate how LBP antibodies enable researchers to dissect specific molecular interactions within the LPS recognition pathway.

What types of LBP antibodies are available for research, and how do they differ?

Research-grade LBP antibodies are available in several formats with distinct characteristics:

Antibody TypeHost SpeciesApplicationsSpecies ReactivityAdvantages
PolyclonalRabbit, SheepWB, IHC, IF, IP, ELISAHuman, Mouse, Rat, othersBroad epitope recognition, robust signal
Monoclonal [N3C3]VariousWB, IHC-pHumanHigh specificity, reduced background
RecombinantVariousWB, ELISA, IP, FAHuman, other speciesConsistent performance, renewable source

Polyclonal antibodies recognize multiple epitopes and are particularly useful for detecting proteins in various applications, while monoclonal antibodies target specific epitopes, providing higher specificity but potentially lower sensitivity . The choice between these depends on experimental requirements, with polyclonals preferred for initial detection and monoclonals for highly specific targeting of LBP domains with known functions . Based on immunogen design, different antibodies target specific regions of LBP, including N-terminal, middle region, and C-terminal epitopes, allowing researchers to investigate domain-specific functions .

What are the optimal protocols for using LBP antibodies in Western blotting?

For effective Western blot detection of LBP, researchers should implement the following methodological considerations:

  • Sample preparation: For tissue lysates, particularly liver tissue where LBP is predominantly synthesized, use RIPA buffer supplemented with protease inhibitors. Tissue samples typically yield more robust signals than cell lines due to LBP's higher expression in certain tissues .

  • Antibody dilution optimization: Most anti-LBP antibodies perform optimally in Western blot applications at dilutions between 1:500-1:2000. For example, Proteintech's 11836-1-AP antibody is recommended at 1:500-1:2000, while R&D Systems' anti-human LBP can be used at 0.2-1.0 μg/mL .

  • Blocking and incubation conditions: Use 5% non-fat milk or BSA in TBST for blocking (1 hour at room temperature), followed by primary antibody incubation overnight at 4°C, which significantly improves signal-to-noise ratio.

  • Detection considerations: LBP typically appears between 53-67 kDa under reducing conditions, with potential variation due to glycosylation and other post-translational modifications. For mouse LBP, bands are often detected at approximately 65 kDa .

  • Loading controls: When comparing LBP expression across different conditions, appropriate loading controls (β-actin for cellular extracts, transferrin for serum samples) must be used due to potential variation in LBP expression during inflammatory conditions.

For validation of specificity, recombinant LBP protein can serve as a positive control, as demonstrated in studies using purified recombinant proteins .

How should LBP antibodies be used in immunohistochemistry to study tissue distribution?

Immunohistochemical detection of LBP requires careful methodological considerations:

  • Sample fixation and processing: Formalin-fixed, paraffin-embedded (FFPE) tissues provide optimal morphological preservation while maintaining LBP antigenicity. Fresh-frozen sections may offer higher sensitivity but poorer morphology.

  • Antigen retrieval methods: Heat-induced epitope retrieval using either citrate buffer (pH 6.0) or TE buffer (pH 9.0) has proven effective for LBP detection. For optimal results with antibody ab231612, TE buffer (pH 9.0) is recommended, while for other antibodies, citrate buffer may be preferred .

  • Antibody concentration and incubation: For FFPE tissues, a concentration range of 5.0-10 μg/mL is typically effective for polyclonal antibodies, with overnight incubation at 4°C enhancing specific binding while reducing background .

  • Detection systems: DAB (3,3'-diaminobenzidine) chromogen detection systems are commonly used, providing stable and permanent staining suitable for archival specimens. Biotinylated secondary antibodies with streptavidin-HRP complexes offer signal amplification for lower-expressing tissues .

  • Controls: Including both positive controls (human or mouse liver tissue, which express high levels of LBP) and negative controls (antibody diluent without primary antibody) is essential for validating staining specificity .

LBP shows distinct tissue distribution patterns, with strongest expression in hepatocytes, moderate expression in epithelial cells of kidney and lung, and variable expression in inflammatory cells, informing experimental design and interpretation .

What approaches are recommended for validating LBP antibody specificity?

Rigorous validation of LBP antibody specificity is crucial for generating reliable research data. Multiple complementary approaches should be employed:

  • Recombinant protein controls: Testing antibodies against purified recombinant LBP protein provides a direct assessment of binding capacity and specificity. Comparing reactivity against standard curves of purified recombinant LBP establishes antibody sensitivity, as demonstrated in studies quantifying LBP in mouse splenic B-cells .

  • Genetic validation approaches: Utilizing tissues or cells from LBP knockout models serves as the gold standard for specificity. Studies have implemented this strategy by comparing antibody reactivity in wild-type versus LBP gene-disrupted mice .

  • Peptide competition assays: Pre-incubating the antibody with the immunizing peptide should abolish specific binding. This method has been used to validate epitope-specific binding of antibodies targeting different LBP domains .

  • Multiple antibody concordance: Using independent antibodies targeting different epitopes of LBP should yield consistent detection patterns. Research has shown that antibodies recognizing distinct regions (N-terminal, middle, and C-terminal) can be used to cross-validate findings .

  • Species cross-reactivity assessment: Systematic testing across multiple species (human, mouse, rat, bovine) establishes the evolutionary conservation of recognized epitopes and informs appropriate experimental models. For example, limited cross-reactivity (less than 10%) between human and mouse LBP has been observed with certain antibodies .

Documented validation data from these approaches should be incorporated into research protocols and publications to support result interpretation and reproducibility.

How can researchers address variable LBP detection across different experimental systems?

Researchers frequently encounter variability in LBP detection, which can be systematically addressed through the following strategies:

  • Understanding expression dynamics: LBP is an acute-phase protein with dramatically variable expression levels (5-10 μg/ml in normal serum to 200 μg/ml during acute-phase response) . This physiological variation necessitates careful timing of experiments and consideration of inflammatory status. Researchers should measure baseline levels and document any potential inflammatory stimuli.

  • Species-specific considerations: Antibody performance often varies across species despite sequence homology. For example, antibodies generated against human LBP may show less than 10% cross-reactivity with mouse LBP . When working with different species, validation using species-specific positive controls is essential.

  • Tissue/cell-specific optimization: LBP expression varies significantly across tissues, with highest expression in liver and lower expression in other tissues. Cell-line detection often requires optimization:

    • For hepatocyte-derived cells: Standard protocols typically yield robust signals

    • For non-hepatic cells: Sample concentration or immunoprecipitation prior to Western blotting may be necessary

    • For serum/plasma: Dilution series testing is essential due to variable concentration ranges

  • Post-translational modification impact: LBP undergoes glycosylation and other modifications that can affect antibody binding and apparent molecular weight (observed ranges of 53-67 kDa) . Deglycosylation experiments may help resolve inconsistent detection patterns.

  • Detection system optimization: If signal variability persists, systematic comparison of different secondary antibodies, detection substrates, and signal amplification methods can identify optimal combinations for specific experimental systems.

Importantly, researchers should maintain detailed records of experimental conditions when LBP detection varies, as these variations may themselves reflect biologically meaningful differences in LBP biology across systems.

What factors might contribute to unexpected molecular weight observations for LBP in Western blots?

Researchers commonly observe variations in LBP's apparent molecular weight across different experimental systems. Several factors contribute to these discrepancies that should be considered during data interpretation:

  • Post-translational modifications: LBP undergoes significant glycosylation, which can increase its apparent molecular weight beyond the calculated 53.4 kDa. Observed weights commonly range from 53-67 kDa in reducing conditions . In mouse ovary tissue, a specific band has been detected at approximately 65 kDa .

  • Species-specific variations: Human LBP may migrate differently than mouse or rat LBP due to species-specific modifications. For instance, bovine LBP shows multiple potential predicted band sizes (37, 50, 53, 55, 57, 60, and 80 kDa) , reflecting possible splice variants or differential processing.

  • Sample preparation effects: Reducing versus non-reducing conditions significantly impact LBP migration patterns. Heat denaturation temperature and duration can also affect observed molecular weights by altering protein conformation and modification status.

  • Tissue-specific processing: LBP from different tissues may undergo differential post-translational processing. While liver-derived LBP is well-characterized (showing bands at 53-60 kDa) , LBP from other tissues may exhibit altered migration patterns.

  • Partial proteolysis: Endogenous proteases or sample preparation artifacts can generate LBP fragments with lower molecular weights. Including protease inhibitors during sample preparation is crucial for consistent results.

When encountering unexpected molecular weights, researchers should compare results with both the calculated molecular weight (53.4 kDa) and the empirically observed ranges from the literature, while documenting experimental conditions that may influence these variations.

How should researchers interpret and reconcile conflicting results from LBP antibody experiments?

When faced with conflicting results from LBP antibody experiments, researchers should implement a systematic approach to resolve discrepancies:

  • Antibody epitope considerations: Different antibodies target distinct regions of LBP, which may be differentially accessible depending on experimental conditions. For example, studies using knockout mice have yielded discordant results regarding LBP's role in endotoxemia, potentially due to differences in the epitopes targeted by neutralizing antibodies . Map the epitopes recognized by each antibody to identify potential conformational dependencies.

  • Experimental context variations: LBP function is highly context-dependent, with its role varying based on:

    • LPS concentration (enhancement of responses at low concentrations versus potential inhibition at high concentrations)

    • Presence of other LPS-binding proteins in the experimental system

    • Cell/tissue type being examined

    • Timing of analysis relative to LPS stimulation

  • Methodological reconciliation approach:

    • Directly compare antibodies under identical conditions

    • Classify antibodies by functional effect (as demonstrated in studies identifying three classes of anti-LBP mAbs with distinct functional properties)

    • Utilize complementary detection methods (e.g., mass spectrometry) to verify identity of detected proteins

  • Dynamic regulation assessment: LBP binding to targets can be rapidly altered by stimulation. For example, ChIP assays demonstrated that LBP-1a binding to S regions decreased significantly 24-48 hours after LPS stimulation, despite increased protein levels . Time-course experiments are essential for capturing such dynamics.

  • Physiological versus pathological contexts: Reconcile findings by distinguishing between normal physiological conditions and inflammatory/pathological states, as LBP functions differently across these contexts .

Research on murine LBP illustrates this approach, where apparently contradictory results from gene knockout studies were resolved through careful characterization of monoclonal antibodies with different functional properties .

How can LBP antibodies be utilized to investigate mechanisms of endotoxin tolerance and sepsis?

LBP antibodies offer sophisticated tools for dissecting the molecular mechanisms underlying endotoxin tolerance and sepsis:

  • Temporal dynamics of LBP-mediated signaling: By using neutralizing antibodies targeting different functional domains of LBP, researchers can establish temporal windows during which LBP activity is critical for progression to sepsis. Studies have demonstrated that class 1 and class 2 mAbs (blocking LPS binding to LBP or LPS/LBP binding to CD14, respectively) protect mice from lethal endotoxemia , suggesting early intervention points.

  • Cell-specific contributions to tolerance: Combining LBP antibodies with cell-specific markers in multiparameter flow cytometry or imaging can reveal:

    • Which cell populations depend on LBP for LPS recognition during tolerance development

    • How LBP-mediated signaling differs between tolerant and non-tolerant cells

    • Whether cellular LBP interactions change during tolerance induction

  • Interventional approaches: Beyond mechanistic studies, LBP antibodies hold therapeutic potential. Research has shown that blockade of LBP activity with polyclonal antibodies protects mice from experimental endotoxemia . This provides a foundation for developing targeted interventions at specific stages of sepsis progression.

  • Molecular interaction mapping: ChIP analyses using LBP antibodies have revealed that LBP binding to genomic regions changes dynamically following stimulation, with binding to Sμ and Sα regions decreasing 24-48 hours post-LPS stimulation . Similar approaches can map how LBP interacts with chromatin during tolerance development.

  • LBP modification monitoring: Phosphorylation or other post-translational modifications may regulate LBP activity during endotoxemia and tolerance. Developing and applying modification-specific antibodies can reveal how these changes correlate with altered LBP function in sepsis progression.

These applications position LBP antibodies as critical tools for understanding the transition from protective inflammatory responses to pathological sepsis and for developing targeted therapeutic interventions.

What role do LBP antibodies play in investigating the connections between gut microbiota, endotoxemia, and inflammatory disorders?

LBP antibodies serve as sophisticated tools for investigating the complex relationships between intestinal barrier function, microbial translocation, and systemic inflammation:

  • Quantifying microbial translocation: LBP levels serve as a reliable biomarker for chronic microbial translocation and low-grade endotoxemia. Using validated LBP antibodies in ELISA or Western blot approaches allows researchers to quantify this process across various disease models and clinical samples . Studies have employed this strategy to correlate gut barrier dysfunction with systemic inflammation in conditions including inflammatory bowel disease, alcoholic liver disease, and metabolic disorders.

  • Tissue-specific LBP expression dynamics: Immunohistochemistry with LBP antibodies can reveal altered expression patterns in intestinal epithelium, liver, and other tissues following dysbiosis or barrier disruption. This approach has identified previously unknown sites of LBP production that may contribute to local inflammatory responses .

  • Mechanistic intervention studies: By combining LBP neutralizing antibodies with models of gut barrier dysfunction, researchers can determine:

    • Whether LBP is necessary for translating microbial signals into inflammatory responses

    • If different microbial products utilize LBP-dependent or -independent recognition pathways

    • How therapeutic manipulation of the microbiome affects LBP-mediated signaling

  • Cell-specific responses to microbial products: Using flow cytometry with LBP antibodies alongside cellular markers allows mapping of which cell populations engage with LBP-bound microbial products across different tissue compartments. This has revealed cell-specific responses that may explain varied outcomes of endotoxemia .

  • Interspecies variation in LBP function: Comparative studies using antibodies reactive across species (human, mouse, rat) can identify conservation and divergence in LBP biology relevant to translating findings between model organisms and humans . Such comparisons have highlighted important differences in LBP concentration dynamics that impact experimental design.

These applications position LBP antibodies as essential tools for understanding how microbial components from the intestinal microbiota initiate and sustain systemic inflammatory processes in diverse pathological conditions.

How can researchers integrate LBP antibody approaches with advanced genomic and proteomic technologies?

Integration of LBP antibody methodologies with cutting-edge omics approaches enables multidimensional insights into LBP biology:

  • ChIP-seq applications: Building on established ChIP protocols for LBP family proteins , ChIP-seq can map genome-wide LBP-1 binding sites during inflammatory responses. This approach has already identified binding to specific immunoglobulin switch regions in B-cells and revealed dynamic binding patterns following LPS stimulation . Extending this to whole-genome analysis could identify previously unknown regulatory targets.

  • Proximity-dependent labeling approaches: Integrating LBP antibodies with BioID or APEX2 proximity labeling systems allows comprehensive mapping of LBP protein interactomes under different conditions. This strategy can reveal:

    • How LBP interaction networks differ between normal and inflammatory states

    • Cell-type specific binding partners that explain differential responses

    • Dynamic changes in protein associations during endotoxin tolerance development

  • Single-cell proteomics integration: Combining LBP antibody staining with mass cytometry (CyTOF) or single-cell westerns enables analysis of LBP expression and modification at single-cell resolution. This approach can identify previously unrecognized cellular heterogeneity in LBP responses that may explain variable outcomes in sepsis and inflammatory disorders.

  • Spatial proteomics applications: Multiplex immunofluorescence or imaging mass cytometry using validated LBP antibodies alongside other markers can map the spatial distribution of LBP relative to its binding partners and effector molecules across tissue microenvironments. This spatial context is crucial for understanding LBP function in complex organs like liver and intestine.

  • Systems biology integration: Data from LBP antibody-based experiments can be integrated with transcriptomics, proteomics, and metabolomics datasets to construct comprehensive network models of LBP-mediated responses. This integrative approach has revealed unexpected connections between LBP signaling and metabolic regulation during inflammatory responses.

These advanced applications transform LBP antibodies from simple detection tools into sophisticated probes for systems-level analysis of host-microbe interactions and inflammatory regulation, advancing our understanding of sepsis, endotoxemia, and related disorders.

What are the current limitations in LBP antibody research and how might they be addressed?

Current LBP antibody research faces several limitations that warrant methodological refinements:

  • Antibody specificity challenges: Despite commercial availability of over 400 LBP antibodies across 29 suppliers , rigorous validation data remains inconsistent. Researchers should:

    • Implement comprehensive validation including knockout/knockdown controls

    • Establish standardized reporting of epitope information and cross-reactivity profiles

    • Develop community-based antibody validation repositories specific to LBP research

  • Functional epitope targeting: Most available antibodies lack precise functional characterization. The field would benefit from systematic classification of antibodies based on their effects on LBP functions (e.g., LPS binding, CD14 interaction), similar to the three functional classes identified in monoclonal antibody studies .

  • Post-translational modification detection: Limited tools exist for detecting and quantifying LBP modifications that likely regulate its function. Development of modification-specific antibodies (phospho-LBP, glyco-LBP) would enable deeper mechanistic insights.

  • Species cross-reactivity limitations: Inconsistent cross-reactivity profiles complicate translation between model organisms and human studies. Systematic characterization of epitope conservation and development of broadly reactive antibodies would facilitate comparative studies.

  • Tissue-specific LBP variant detection: Current antibodies may not distinguish potential tissue-specific LBP variants or isoforms. Epitope mapping combined with RNA-seq data could guide development of isoform-selective antibodies.

Addressing these limitations requires collaborative efforts between antibody developers, LBP researchers, and organizations focused on antibody validation standards. Implementation of systematic validation workflows and detailed reporting of antibody characteristics in publications would significantly advance the field.

What emerging research directions may benefit most from LBP antibody technologies?

Several cutting-edge research areas are poised to benefit substantially from advanced LBP antibody applications:

  • Microbiome-immune interface research: As interest in microbiome-host interactions grows, LBP antibodies will be crucial for understanding how microbial signals are interpreted by the host immune system. Specifically, research into how different microbial communities modulate LBP function in intestinal barrier diseases represents a high-priority area where antibody-based detection of LBP-microbe interactions could provide mechanistic insights .

  • Single-cell immunology of sepsis: Heterogeneity in cellular responses to endotoxin remains poorly understood. Combining LBP antibodies with single-cell technologies can reveal how individual cells within populations differentially respond to LBP-LPS complexes, potentially explaining variable clinical outcomes in sepsis patients.

  • Tissue-resident immunity in non-classical sites: Recent discovery of tissue-specific immune compartments suggests LBP may function differently across tissues. Immunohistochemistry and tissue-clearing techniques with LBP antibodies could map previously unrecognized sites of LBP activity beyond classical immune organs .

  • Inflammatory mechanisms in metabolic diseases: Mounting evidence connects low-grade endotoxemia to metabolic disorders. LBP antibodies enable investigation of how subclinical LPS exposure through altered gut permeability contributes to insulin resistance, non-alcoholic steatohepatitis, and cardiovascular disease .

  • Therapeutic antibody development: Building on protective effects observed with neutralizing LBP antibodies in experimental endotoxemia , development of therapeutic-grade humanized antibodies targeting specific LBP functional domains represents a promising approach for sepsis intervention.

Each of these research directions would benefit from continued refinement of LBP antibody technologies and validation methodologies, positioning these reagents as essential tools for addressing fundamental questions at the intersection of microbial recognition and inflammatory regulation.

What standardization approaches could improve reproducibility in LBP antibody research?

To enhance reproducibility and facilitate cross-study comparisons in LBP antibody research, implementation of the following standardization approaches is recommended:

  • Comprehensive validation reporting framework: Researchers should adopt standardized reporting of validation data including:

    • Epitope information with amino acid residue specifications

    • Validation methods employed (Western blot, IP-MS, knockout controls)

    • Quantitative cross-reactivity profiles across species

    • Functional effects on LBP activities

    Such comprehensive reporting would allow other researchers to select appropriate antibodies for specific applications and experimental systems.

  • Reference material establishment: Development and distribution of:

    • Recombinant LBP protein standards for calibration across studies

    • Standardized positive control lysates from relevant tissues/cell types

    • Validated LBP knockout/knockdown cell lines as negative controls

  • Methodological protocol standardization: Creating consensus protocols for:

    • Optimal fixation and antigen retrieval for immunohistochemistry

    • Sample preparation guidelines for Western blotting to address molecular weight variations

    • Standardized dilution series for immunoassay calibration

    • Detailed ChIP protocols optimized for LBP binding studies

  • Functional classification system: Implementing a standardized functional classification system for LBP antibodies similar to the three-class system described for monoclonal antibodies (LPS binding inhibitors, CD14 interaction blockers, non-neutralizing binders) . This would enable researchers to select antibodies based on desired functional effects.

  • Central repository for validation data: Establishing a community resource database where researchers can:

    • Submit independent validation data for commercial and custom antibodies

    • Access standardized protocols optimized for specific applications

    • Compare antibody performance across experimental systems

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 2025 TheBiotek. All Rights Reserved.