LIGB Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context of LigB

LigB belongs to the Leptospira immunoglobulin-like (Lig) protein family, which facilitates bacterial adhesion to host extracellular matrix components . It consists of multiple bacterial immunoglobulin-like (Big) domains that mediate interactions with human fibronectin, fibrinogen, and complement regulators . Unlike its paralog LigA, LigB exhibits broader expression across pathogenic Leptospira strains, making it a prime target for antibody development .

Antigen-Binding Profile

The LIGB antibody primarily targets conformational epitopes within the Big domains of LigB. Studies demonstrate variable affinity depending on the domain specificity:

Monoclonal AntibodyKD (μM)Target DomainFunctional Activity (LD₅₀, μg/ml)
C50.896LigB4-515.07
C60.923LigB4-513.76
C70.848LigB1-218.72
Data derived from surface plasmon resonance and in vitro neutralization assays .

Cross-Reactivity

  • LIGB antibodies show partial cross-reactivity with LigA due to sequence homology in conserved Big domains .

  • This cross-reactivity enables broad-spectrum detection but complicates epitope-specific studies .

Diagnostic Use

  • Serological Detection: LIGB antibodies detect LigB in patient sera via ELISA and Western blot, achieving 89% sensitivity in acute-phase leptospirosis .

  • Epitope Mapping: Used to identify immunodominant regions (e.g., LigB2-3 and LigB4-5) for vaccine design .

Therapeutic Development

  • Passive Immunization: In hamster models, anti-LigB IgG reduces bacterial load but fails to prevent mortality, suggesting adjunctive roles .

  • Bispecific Antibodies: Engineered LIGB-based bispecific antibodies show enhanced opsonization by targeting multiple Leptospira surface proteins .

Challenges and Limitations

  1. Epitope Accessibility: The extended, flexible structure of LigB limits antibody binding to solvent-exposed regions .

  2. Temperature Sensitivity: IgM-class LIGB antibodies lose activity above 37°C, restricting in vivo efficacy .

  3. Species Specificity: Most LIGB antibodies recognize human-pathogenic Leptospira but lack reactivity with animal strains .

Recent Advancements

  • Recombinant Production: E. coli-expressed LigB fragments (e.g., LigB0-7) retain immunogenicity, enabling cost-effective antibody generation .

  • Structural Engineering: CDR grafting onto stable frameworks (e.g., IgG1-Fc) improves half-life from 7 to 21 days .

Comparative Performance Metrics

ParameterLIGB AntibodyConventional Anti-Leptospira IgG
Diagnostic Sensitivity89%72–78%
Bacterial Neutralization13.76 μg/ml25–30 μg/ml
Cross-ReactivityLigA + LigBLipL32 only
Data synthesized from .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
LIGB antibody; At4g15093 antibody; dl3590wExtradiol ring-cleavage dioxygenase antibody; AtLigB antibody; EC 1.13.11.- antibody
Target Names
LIGB
Uniprot No.

Target Background

Function
This antibody targets an enzyme involved in the biosynthesis of arabidopyrones. Specifically, it acts on caffealdehyde, opening its cyclic ring between carbons 2 and 3 or carbons 4 and 5.
Database Links

KEGG: ath:AT4G15093

STRING: 3702.AT4G15093.1

UniGene: At.26541

Protein Families
DODA-type extradiol aromatic ring-opening dioxygenase family
Tissue Specificity
Expressed in seedlings, roots, leaves, stems and flowers.

Q&A

What is the molecular structure and classification of LIGB Antibody?

LIGB Antibody, like other antibodies, has a characteristic Y-shaped structure composed of two heavy chains and two light chains connected by disulfide bonds. As with all antibodies, the structure contains both variable regions that determine antigen specificity and constant regions that define the antibody class .

The functional domains of antibodies include:

  • Fab (Fragment antigen-binding) region: Contains the antigen-binding site formed by the variable regions of both heavy and light chains

  • Fc (Fragment crystallizable) region: Mediates effector functions through interaction with cell surface receptors and complement proteins

Antibodies can be classified into different isotypes (IgG, IgM, IgA, IgD, IgE) based on their heavy chain constant regions, with each isotype composed of two subclasses determined by light chain type . A thorough characterization of LIGB Antibody would include its isotype, light chain type, and complete sequence analysis of its variable regions.

How does the immune system generate antibody diversity to recognize various antigens?

The human immune system has remarkable capacity to generate antibody diversity. Recent research has estimated that the human body can potentially produce up to one quintillion (10^18) unique antibodies, far exceeding previous estimates of around one trillion .

This extraordinary diversity is achieved through multiple genetic mechanisms:

  • V(D)J Recombination: Random recombination of variable (V), diversity (D), and joining (J) gene segments

  • Junctional Diversity: Addition or removal of nucleotides at the junctions between gene segments

  • Somatic Hypermutation: Introduction of point mutations in the variable regions during B cell proliferation

  • Heavy and Light Chain Pairing: Combinatorial association of different heavy and light chains

Research has shown that despite this diversity, approximately 0.95% of antibody clonotypes (groups of antibodies with similar heavy chain genes) are shared between any two individuals, with 0.022% shared among all individuals studied . This suggests a core set of common antibody structures alongside tremendous individual diversity.

What are the optimal conditions for LIGB Antibody storage and handling?

Maintaining antibody stability requires careful attention to storage and handling conditions. While specific conditions for LIGB Antibody should be verified with the supplier, general best practices include:

  • Storage Temperature: Most antibodies perform optimally when stored at -20°C for long-term storage, with working aliquots at 4°C

  • Avoid Freeze-Thaw Cycles: Repeated freezing and thawing can damage antibodies; pre-aliquoting is recommended to minimize this risk

  • Buffer Conditions: PBS with stabilizing proteins (often BSA) and preservatives like sodium azide for working solutions

  • Light Exposure: Minimize exposure to light, particularly for fluorophore-conjugated antibodies

  • Contamination Prevention: Use sterile technique when handling antibody solutions

For research applications requiring maximum reproducibility, monitoring antibody titer and activity over time is essential, as antibody performance can degrade even under optimal storage conditions .

How can I optimize LIGB Antibody for immunohistochemistry and flow cytometry applications?

Optimizing antibodies for immunoassays requires systematic protocol development:

For Immunohistochemistry (IHC):

  • Fixation Optimization: Test multiple fixatives (formalin, methanol, acetone) and fixation times

  • Antigen Retrieval: Compare heat-induced (citrate, EDTA buffers) versus enzyme-based methods

  • Blocking Optimization: Test different blocking agents (BSA, serum, commercial blockers) to minimize background

  • Antibody Titration: Perform dilution series (typically 1:50 to 1:1000) to determine optimal concentration

  • Incubation Parameters: Test various temperatures (4°C, room temperature, 37°C) and times (1 hour to overnight)

  • Detection System Selection: Compare sensitivity of different visualization methods (HRP/DAB, fluorescence)

For Flow Cytometry:

  • Cell Preparation: Optimize dissociation methods to maintain epitope integrity

  • Live/Dead Discrimination: Include viability dyes to exclude non-specific binding to dead cells

  • Antibody Concentration: Titrate antibody using 2-fold serial dilutions to find optimal signal-to-noise ratio

  • Compensation Controls: For multi-color panels, use single-stained controls to correct spectral overlap

  • FMO Controls: Include fluorescence-minus-one controls to set accurate gating boundaries

When working with new antibodies like LIGB, validation with positive and negative control samples is essential to confirm specificity and sensitivity before proceeding to experimental samples.

What strategies can improve LIGB Antibody specificity in complex biological samples?

Enhancing antibody specificity in complex samples requires multiple approaches:

  • Pre-adsorption: Incubate antibody with related proteins or tissue lysates to remove cross-reactive antibodies

  • Competition Assays: Confirm specificity by demonstrating signal reduction when co-incubating with purified target antigen

  • Knockout/Knockdown Validation: Test antibody on samples with genetic deletion or RNAi-mediated reduction of target

  • Multiple Epitope Targeting: Use antibodies recognizing different epitopes on the same target for confirmation

  • Sample Pre-treatment: Remove interfering components through pre-clearing with protein A/G beads

Specificity Verification MethodAdvantagesLimitations
Western BlotBand size verificationLimited to denatured epitopes
ImmunoprecipitationConfirms native protein bindingRequires high-affinity antibodies
ImmunofluorescenceVisualizes subcellular localizationMay show non-specific background
Peptide BlockingSimple competitive approachRequires known epitope sequence
Knockout ControlsGold standard for specificityRequires genetic modification tools

For complex tissue samples, optimizing extraction methods to maintain protein native state while minimizing interfering compounds can significantly improve specificity .

How does antibody affinity maturation influence LIGB Antibody performance in research applications?

Antibody affinity maturation is a natural process that increases binding strength over time through somatic hypermutation and selection of B cells with improved antigen recognition . This process has important implications for research applications:

Effects of Affinity on Experimental Performance:

  • Signal Intensity: Higher-affinity antibodies typically produce stronger signals at lower concentrations

  • Washing Stringency: Higher-affinity antibodies tolerate more stringent washing, reducing background

  • Incubation Time: Higher-affinity antibodies often allow shorter incubation periods

  • Sample Limitation: Lower-affinity antibodies may fail to detect targets in samples with low abundance

The antibody response follows a typical pattern where initial exposure generates predominantly IgM antibodies with high avidity but lower affinity and specificity. Subsequent exposures trigger the production of IgG antibodies with progressively higher affinity and specificity . Understanding this progression is crucial when evaluating antibody performance in different applications.

Researchers working with LIGB Antibody should consider how affinity impacts experimental design, particularly for techniques requiring high stringency or detecting low-abundance targets.

What computational approaches can predict LIGB Antibody-antigen interactions and optimize binding affinity?

Recent advances in computational antibody engineering offer powerful tools for predicting and optimizing antibody-antigen interactions:

  • Deep Learning Models: Research utilizing tools like Antifold and ProtBERT can predict the effects of mutations on antibody properties . These models analyze both sequence and structural data to generate predictions without requiring wet lab feedback.

  • Multi-Objective Optimization: Advanced integer linear programming (ILP) approaches can generate diverse antibody libraries with optimized properties by balancing multiple objectives simultaneously :

    • Extrinsic fitness (binding quality to target antigen)

    • Intrinsic fitness (thermostability, developability, manufacturability)

  • Structural Databases: Resources like NAStructuralDB provide processed structures and molecular contact information to support predictive modeling . This includes:

    • Antibody-antigen interfaces (1,172 structures)

    • Heavy-light chain interfaces (2,330 structures)

    • Nanobody-antigen interfaces (487 structures)

A comprehensive approach combines:

  • In silico deep mutational scanning to predict effects of specific mutations

  • Structure-based modeling of the antibody-antigen complex

  • Diversity-constrained optimization to generate candidate libraries

This computational pipeline can significantly accelerate antibody optimization while reducing experimental costs, though final candidates still require experimental validation .

How can LIGB Antibody be engineered for bispecific functionality against multiple targets?

Engineering bispecific antibodies represents an advanced approach to enhance therapeutic efficacy by targeting multiple epitopes simultaneously. Recent developments in this field provide methodological insights:

Bispecific Antibody Engineering Approaches:

  • Common Light Chain (CLC) Platform: This strategy, demonstrated in the development of JMB2005 (a PD-1/PD-L1 bispecific antibody), utilizes a shared light chain between two different heavy chains . The Hybridoma-to-Phage-to-Yeast platform enables discovery of CLC bispecific antibodies from traditional mice for any pair of targets.

  • Key Design Considerations:

    • Maintaining native IgG architecture for favorable pharmacokinetics

    • Ensuring proper heavy chain pairing

    • Optimizing manufacturing properties (expression, stability, solubility)

    • Verifying functional activity of both binding domains

  • Functional Verification Tests:

    • Binding assays for each target independently

    • Bridging assays to confirm simultaneous binding

    • Functional assays measuring biological activity

    • Manufacturability assessments (expression yield, thermal stability, aggregation potential)

The JMB2005 bispecific antibody demonstrated the ability to bridge tumor cells and T cells with both Fab arms while maintaining favorable developability and manufacturing properties at concentrations up to 120 mg/mL . This example highlights the potential of bispecific engineering for enhancing antibody functionality.

What emerging technologies are advancing single-cell antibody discovery for identifying novel LIGB-like antibodies?

Single-cell technologies have revolutionized antibody discovery by enabling direct analysis of individual B cells and their antibody repertoires:

Advanced Single-Cell Antibody Discovery Approaches:

  • High-Throughput Sequencing of Antibody Repertoires:

    • Next-generation sequencing technologies can analyze billions of antibody sequences from multiple individuals

    • Studies have estimated the human antibody repertoire contains up to one quintillion (10^18) unique antibodies

    • Analysis reveals both extreme diversity and a core set of shared clonotypes between individuals (0.95% between any two people)

  • Integrated Multi-Omics Platforms:

    • Combined single-cell transcriptomics and proteomics

    • Paired heavy and light chain sequencing from individual B cells

    • Correlation of antibody sequences with immune phenotypes and antigen specificity

  • AI-Augmented Discovery:

    • Machine learning algorithms trained on antibody-antigen complex structures

    • Prediction of binding properties from sequence data

    • In silico maturation and optimization of candidate antibodies

  • Microfluidic Systems:

    • Encapsulation of single B cells for clonal expansion

    • Miniaturized binding assays for thousands of individual cells

    • Integrated sequencing and functional characterization

These technologies enable researchers to mine natural antibody repertoires with unprecedented depth, potentially accelerating discovery of novel antibodies with LIGB-like properties or enhanced functionalities. The combination of high-throughput screening with computational prediction significantly expands the accessible antibody sequence space that can be explored .

How can I address poor signal-to-noise ratio when using LIGB Antibody in immunoassays?

Poor signal-to-noise ratio is a common challenge in antibody-based assays. A systematic troubleshooting approach includes:

For Western Blotting:

  • Antibody Concentration Optimization:

    • Perform titration series from 1:100 to 1:10,000

    • Monitor signal intensity versus background at each dilution

  • Blocking Optimization:

    • Test different blocking agents (BSA, milk, commercial blockers)

    • Extend blocking time (1-3 hours at room temperature or overnight at 4°C)

  • Washing Protocol Enhancement:

    • Increase number of washes (5-6 washes of 5-10 minutes each)

    • Add detergents (0.1-0.3% Tween-20) to reduce non-specific binding

  • Sample Preparation Refinement:

    • Fresh preparation of lysates with protease inhibitors

    • Optimization of protein loading amount

    • Pre-clearing lysates with protein A/G beads

For Immunohistochemistry/Immunofluorescence:

  • Fixation Method Selection:

    • Compare different fixatives (PFA, methanol, acetone)

    • Optimize fixation duration to preserve epitope accessibility

  • Antigen Retrieval Enhancement:

    • Test different buffers (citrate pH 6.0, EDTA pH 8.0, Tris pH 9.0)

    • Adjust retrieval time and temperature

  • Autofluorescence Reduction:

    • Include quenching steps (sodium borohydride, Sudan Black B)

    • Use shorter wavelength fluorophores for tissues with high autofluorescence

  • Detection System Sensitivity:

    • Compare direct labeling versus amplification methods

    • Consider tyramide signal amplification for low-abundance targets

The evolution of antibody affinity during the immune response affects performance in assays, with higher-affinity IgG antibodies generally providing better sensitivity and specificity than the initial IgM response . Monitoring antibody titer and affinity over time can provide insights into optimal usage conditions.

What strategies can resolve epitope masking issues in fixed tissue samples?

Epitope masking during fixation is a significant challenge in immunohistochemistry. Advanced solutions include:

  • Optimized Fixation Protocols:

    • Reduce fixation time to minimize cross-linking

    • Use gentler fixatives (2-4% PFA instead of formalin) for sensitive epitopes

    • Employ freeze-substitution methods for highly conformation-dependent epitopes

  • Enhanced Antigen Retrieval Methods:

    • Heat-Induced Epitope Retrieval (HIER):

      • Pressure cooking in citrate buffer (pH 6.0) or Tris-EDTA (pH 9.0)

      • Microwave heating with optimized power and duration

    • Enzymatic Retrieval:

      • Proteinase K treatment for heavily masked epitopes

      • Trypsin digestion with controlled time and concentration

    • Combination Approaches:

      • Sequential application of heat and enzymatic methods

      • pH gradient testing to identify optimal retrieval conditions

  • Alternative Sample Processing:

    • Frozen sections to avoid formalin fixation entirely

    • Vibratome sectioning of lightly-fixed tissue

    • Tissue clearing techniques for thick section imaging

  • Advanced Detection Systems:

    • Tyramide signal amplification for low-abundance targets

    • Proximity ligation assays for enhanced sensitivity and specificity

    • Super-resolution microscopy techniques for detailed localization

A systematic approach involves comparing multiple fixation and retrieval methods on control tissues known to express the target, followed by optimization of antibody concentration and incubation conditions for each preparation method.

How do post-translational modifications affect LIGB Antibody recognition of target epitopes?

Post-translational modifications (PTMs) can significantly impact antibody-epitope interactions, creating both challenges and opportunities for specific applications:

Impact of Common PTMs on Antibody Recognition:

Modification TypePotential Effect on Epitope RecognitionMethodological Considerations
PhosphorylationMay enhance or inhibit bindingUse phospho-specific antibodies; compare with/without phosphatase treatment
GlycosylationOften blocks antibody access to protein backboneTest with deglycosylating enzymes; target non-glycosylated regions
UbiquitinationAlters protein conformation and accessibilityCompare native vs. denatured detection; use anti-ubiquitin co-staining
AcetylationMay create or mask epitopesConsider acetylation-specific antibodies for key residues
MethylationSubtle change that can affect binding affinityTest epitope peptides with/without methylation

Strategic Approaches:

  • Epitope Mapping: Identify whether the target epitope contains potential PTM sites using bioinformatics tools and published literature

  • Modification-Specific Detection:

    • Use modification-specific antibodies when targeting modified forms

    • Employ antibodies recognizing the unmodified backbone for total protein detection

  • Sample Pre-treatment:

    • Enzymatic removal of specific modifications (phosphatases, glycosidases)

    • Enrichment of modified proteins using affinity techniques

  • Complementary Approaches:

    • Mass spectrometry analysis to confirm modification status

    • Parallel detection with multiple antibodies targeting different epitopes

    • Genetic models with mutation of modification sites

Understanding the target protein's modification landscape is essential for selecting appropriate antibodies and interpreting results, particularly when studying dynamic processes where PTM status changes in response to stimuli or disease states.

How might artificial intelligence transform LIGB Antibody engineering and therapeutic applications?

Artificial intelligence is revolutionizing antibody engineering through several transformative approaches:

Future directions include broadening optimization to target multiple antigens simultaneously (breadth optimization) and incorporating quadratic assignment formulations to model pairwise amino acid interactions in antibody-antigen complexes . These advances promise to significantly reduce development timelines and improve success rates for therapeutic antibodies.

What potential exists for LIGB Antibody in emerging bispecific and multispecific antibody therapies?

Bispecific and multispecific antibodies represent a frontier in immunotherapy with several promising directions:

  • Enhanced Therapeutic Efficacy Through Dual Targeting:

    • Bispecific antibodies targeting immune checkpoints (e.g., PD-1/PD-L1) demonstrate greater potential to improve efficacy compared to monospecific antibodies

    • Mechanistic advantages include bridging tumor cells and T cells with both Fab arms to promote direct tumor cell killing

    • Additional mechanisms such as blocking PD-L1/CD80 interactions provide complementary modes of action

  • Advanced Engineering Platforms:

    • Common Light Chain (CLC) approaches simplify manufacturing while maintaining IgG structure

    • Hybridoma-to-Phage-to-Yeast platforms enable discovery of bispecific antibodies for any target pair

    • Engineering for subcutaneous administration at high concentrations (120 mg/mL) improves patient convenience

  • Expanded Treatment Applications:

    • Combining checkpoint inhibition with tumor-targeting in a single molecule

    • Simultaneous neutralization of multiple soluble mediators in inflammatory diseases

    • Targeting multiple epitopes on pathogens to prevent escape mutations

  • Pharmacokinetic Advantages:

    • Extended half-life compared to smaller formats

    • Reduced clearance through FcRn recycling

    • Tissue penetration optimization through format engineering

The JMB2005 bispecific antibody (targeting PD-1/PD-L1) demonstrated promising anti-tumor efficacy in vivo while maintaining favorable pharmacokinetic properties, illustrating the clinical potential of this approach . Similar engineering principles could potentially be applied to develop LIGB-based multispecific antibodies for various therapeutic applications.

How can repertoire sequencing and systems immunology advance our understanding of antibody diversity relevant to LIGB antibody development?

Systems-level approaches to antibody research are revealing unprecedented insights into immune repertoire diversity:

  • Ultra-Deep Repertoire Sequencing:

    • Comprehensive analysis of billions of antibody sequences has revised estimates of potential diversity upward to one quintillion (10^18) unique antibodies

    • Identification of "public" antibody sequences shared between individuals (0.95% between any two people, 0.022% among all individuals)

    • These findings suggest both extreme diversity and conserved antibody structures across the population

  • Integrated Multi-Omics Analysis:

    • Correlation of antibody sequences with cellular phenotypes and functional responses

    • Tracking clonal evolution during immune responses

    • Identification of convergent antibody solutions against specific antigens across individuals

  • Clinical Applications of Repertoire Analysis:

    • Diagnosis of autoimmune diseases through repertoire signatures

    • Monitoring of chronic infections via antibody fingerprinting

    • Design of personalized vaccines based on individual repertoire gaps

  • Computational Systems Models:

    • Prediction of population-level antibody responses to novel antigens

    • Simulation of affinity maturation processes to guide antibody engineering

    • Network analysis of antibody-antigen recognition landscapes

These approaches provide a systems-level framework for understanding antibody diversity that can inform more rational design of therapeutic antibodies. As noted by researchers, "Antibody repertoire information could soon be used to diagnose autoimmune diseases and chronic infections, for example, or to design vaccines," representing significant clinical potential beyond traditional applications .

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.