NERD Antibody

Shipped with Ice Packs
In Stock

Description

Understanding NERD and Antibody Therapies

NSAID-Exacerbated Respiratory Disease (NERD) involves airway inflammation driven by overproduction of cysteinyl leukotrienes (CysLTs) and type 2 eosinophilic inflammation . While no antibody is termed "NERD Antibody," several biologics targeting inflammatory pathways are used in NERD management.

Therapeutic AntibodyTargetMechanismClinical Relevance
DupilumabIL-4RaBlocks IL-4/IL-13 signalingImproves nasal/asthma symptoms in NERD
MepolizumabIL-5Reduces eosinophil countsReduces exacerbations in severe asthma
TezepelumabTSLPInhibits epithelial cell activationUnder investigation for NERD

Antibody Structure and Function in Inflammatory Diseases

Antibodies used in NERD and similar conditions are typically IgG isotypes, which dominate blood and tissue fluids due to their small size and placental transfer ability 5. Their structure includes:

  • Two heavy chains (H) and two light chains (L), forming a Y-shaped molecule with antigen-binding sites at the tips .

  • Constant regions (Fc) mediating effector functions (e.g., complement activation, Fc receptor binding) .

For example, dupilumab is an IgG4 antibody engineered for high-affinity binding to IL-4Ra, preventing IL-4/IL-13 interaction and reducing eosinophilic inflammation .

Research and Development Challenges

Antibody engineering for NERD involves optimizing binding affinity and stability. Key strategies include:

  • Humanization: Replacing non-human residues with human equivalents to minimize immunogenicity .

  • Avidity modulation: Enhancing multivalent binding via bispecific antibodies or Fc engineering .

Recent advancements in computational models (e.g., IgFold and AbMap) enable rapid prediction of antibody structures and binding kinetics, accelerating drug discovery .

Limitations and Future Directions

While antibodies like dupilumab show promise, challenges remain:

  • Heterogeneity: NERD patients exhibit diverse phenotypes (e.g., CRS with/without urticaria), requiring personalized therapies .

  • Cost and access: Therapeutic antibodies are expensive and not universally available.

Future research may focus on nanobodies (single-domain antibodies) for targeted delivery to airway mucosa or bispecific antibodies targeting multiple inflammatory pathways (e.g., IL-5 + IL-4) .

Key Findings from NERD Studies

Study FocusObservationSource
Dupilumab efficacyImproved FEV1 and nasal polyp scores in NERD patients
CysLT overproductionElevated LTE4 levels correlate with airway obstruction severity
Genetic predispositionVariants in HTR2A and TSLP linked to NERD susceptibility

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
NERD antibody; At2g16485/At2g16480/At2g16470 antibody; F16F14Zinc finger CCCH domain-containing protein 19 antibody; AtC3H19 antibody; Protein Needed for RDR2-independent DNA methylation antibody
Target Names
NERD
Uniprot No.

Target Background

Function
This antibody targets a protein that plays a crucial role in integrating RNA silencing and chromatin signaling pathways. Specifically, it's involved in the 21-nucleotide siRNA-dependent DNA methylation pathway on cytosine, leading to transcriptional gene silencing of specific sequences. This protein participates in a chromatin-based RNA silencing pathway encompassing both post-transcriptional gene silencing (PTGS; e.g., RDR1, RDR6, and AGO2) and transcriptional gene silencing (TGS; e.g., siRNA-dependent DNA methylation and histone H3 modifications). It mediates siRNA accumulation at specific chromatin loci and binds to histone H3K4me0 via its PHD domain, maintaining low levels of H3K4 methylation and ensuring gene silencing at specific genomic loci.
Gene References Into Functions
  1. Evidence supporting a central role for this protein in integrating RNA silencing and chromatin signals in transcriptional silencing comes from its demonstrated binding to both histone H3 and AGO2 proteins, and its contribution to siRNA accumulation at a target locus. (PMID: 22940247)
Database Links

KEGG: ath:AT2G16485

STRING: 3702.AT2G16485.1

UniGene: At.40269

Subcellular Location
Nucleus.
Tissue Specificity
Expressed in seedlings, mostly in the vasculature and shoot apices of young seedlings.

Q&A

What is the basic structural composition of antibodies?

Antibodies (immunoglobulins) consist of four polypeptide chains - two identical heavy chains and two identical light chains - connected by disulfide bonds to form a Y-shaped molecule. Each chain contains constant and variable regions, with the variable regions forming the antigen-binding site. The basic antibody structure includes Fab regions (antigen-binding fragments) containing the variable domains and the Fc region (crystallizable fragment) that interacts with cell surface receptors and complement proteins14.

The variable domains contain three complementarity-determining regions (CDRs) that form the antigen-binding pocket, with CDR3 typically showing the highest variability. These structural elements enable the remarkable specificity of antibodies while maintaining conserved effector functions through the constant regions1 .

How do the five major classes of antibodies differ structurally and functionally?

The five major immunoglobulin classes differ in their heavy chain structure, distribution, and functional properties:

Antibody ClassStructureAbundancePrimary LocationKey Functions
IgGMonomerMost abundant (65-70% of serum antibodies)Blood plasmaComplement activation, opsonization, neutralization of toxins and viruses, placental transfer1
IgMPentamer (or monomer)Present during primary immune responseExtracellular fluidsPrimary response to new antigens, efficient complement activation with 10 antigen-binding sites in pentameric form1
IgADimer (or monomer)Predominant in mucosal secretionsMucosal surfaces, secretionsMucosal immunity, prevention of pathogen attachment to epithelial surfaces1
IgDMonomerLow serum levelsB cell surfaceB cell development and activation4
IgEMonomerVery low serum levelsBound to mast cells and basophilsParasitic immunity, allergic reactions4

Each antibody class serves specialized roles in immune protection based on their structural characteristics. For instance, the pentameric structure of IgM provides multiple binding sites that enhance its avidity for antigens during initial exposure, making it a crucial first responder despite its relatively lower affinity compared to IgG14.

What mechanisms determine antibody specificity at the molecular level?

Antibody specificity is determined primarily by the three-dimensional structure of the variable regions, particularly the complementarity-determining regions (CDRs). The CDR3 region typically contributes most significantly to specificity determination due to its high variability and central position in the antigen-binding site 4.

At the molecular level, specificity arises from:

  • Sequence variation in CDRs that creates unique binding pockets

  • Conformational adaptability of the binding site

  • Distribution of charged, polar, and hydrophobic residues that facilitate complementary interactions with antigen epitopes

  • Subtle structural adjustments upon antigen binding that enhance fit (induced fit mechanism)

Recent research indicates that antibody specificity can be modeled computationally by identifying distinct binding modes associated with particular ligands. This approach allows researchers to disentangle multiple binding contributions and design antibodies with customized specificity profiles, either for binding to specific targets or cross-reactivity against multiple related targets .

How can mass spectrometry techniques revolutionize the detection and characterization of monoclonal antibodies?

Mass spectrometry has transformed antibody analysis by providing enhanced sensitivity and specificity compared to traditional electrophoretic methods. The "mass fix" method, developed at Mayo Clinic, has significantly improved the detection of monoclonal proteins (M-proteins) in serum samples3.

The methodology involves:

  • Immunoprecipitation with heavy-chain specific antibodies to isolate specific immunoglobulin classes

  • Release and analysis of light chains using MALDI-TOF (Matrix-Assisted Laser Desorption/Ionization-Time of Flight) mass spectrometry

  • Comparison of kappa and lambda light chain distributions to identify clonal expansions

This technique provides several advantages over traditional techniques:

  • Higher sensitivity for detecting low-level disease in multiple myeloma patients

  • Improved ability to monitor minimal residual disease following treatment

  • Enhanced capacity to detect post-translational modifications that may indicate disease progression

  • Superior specificity when combined with other diagnostic modalities3

Recent studies have demonstrated that high-sensitivity mass spectrometry methods (LC-ESI-MS) for serum analysis may even outperform next-generation sequencing of bone marrow samples in predicting progression-free survival in multiple myeloma patients, highlighting the clinical significance of these advanced analytical approaches3.

What computational approaches enable the design of antibodies with customized specificity profiles?

Modern antibody design leverages sophisticated computational models that integrate experimental data from phage display selections with biophysical principles. These approaches enable researchers to:

  • Identify distinct binding modes associated with specific target ligands

  • Disentangle contributions from multiple epitopes even when they cannot be experimentally isolated

  • Design antibodies with tailored specificity profiles that either target individual ligands with high specificity or exhibit controlled cross-reactivity

A particularly powerful approach combines high-throughput sequencing of antibody libraries with biophysics-informed modeling. This methodology assigns energy functions to different binding modes, allowing researchers to optimize sequences for desired specificity profiles by minimizing energy functions for desired targets while maximizing them for unwanted interactions .

The workflow typically involves:

  • Conducting phage display experiments with antibody libraries against various ligand combinations

  • Sequencing selected antibodies to establish training datasets

  • Building computational models that associate binding modes with specific ligands

  • Optimizing antibody sequences based on the energy functions associated with each mode

  • Experimental validation of computationally designed variants

This approach has been successfully demonstrated with minimal antibody libraries based on a single naïve human V domain with variations in the third complementarity-determining region (CDR3), allowing discrimination between chemically similar ligands even when they cannot be experimentally dissociated from other epitopes present during selection .

How can researchers differentiate between monoclonal antibody production and polyclonal immune responses in clinical samples?

Differentiating between monoclonal antibody production (indicative of plasma cell disorders) and polyclonal immune responses requires sophisticated analytical approaches:

  • Mass Spectrometry-Based Methods:

    • MALDI-TOF analysis reveals distinctive narrow peaks for monoclonal antibodies against the broader polyclonal background

    • The mass distribution of light chains shows characteristic patterns: monoclonal antibodies produce sharp, dominant peaks, while polyclonal responses show broader distributions with typical kappa:lambda ratios (1:1 to 1.5:1 for IgG and IgA, lower for IgM)3

  • Sequential Monitoring:

    • Persistence of identical molecular weight signatures across multiple time points suggests monoclonality

    • Polyclonal responses typically show greater variability or resolution over time

  • Post-Translational Modification Analysis:

    • Examination of peak patterns can reveal post-translational modifications of a primary clone, which must be distinguished from truly unique clones

    • The presence of satellite peaks with defined mass differences often indicates modifications of a monoclonal protein rather than separate clones3

Using these advanced techniques, researchers can achieve significantly higher sensitivity than traditional electrophoretic methods, enabling detection of minimal residual disease in multiple myeloma and other plasma cell disorders. The combination of serum-based mass spectrometry with bone marrow analyses provides complementary information, with mass spectrometry offering particular value in detecting low-level persistence of disease3.

How should researchers optimize phage display experiments for generating antibodies with specific binding profiles?

Optimizing phage display experiments requires careful consideration of several critical parameters:

  • Library Design and Diversity:

    • Strategic variation of complementarity-determining regions (CDRs), particularly CDR3, which contributes most significantly to binding specificity

    • Implementation of systematic amino acid substitutions across targeted positions to generate comprehensive variant libraries

    • Maintenance of high coverage (>40% of theoretical variants) to ensure thorough exploration of sequence space

  • Selection Strategy:

    • Implementation of positive selections against target ligands to identify binding sequences

    • Incorporation of counter-selections against unwanted ligands to eliminate cross-reactive antibodies

    • Adjustment of selection stringency across multiple rounds to balance breadth of capture versus affinity maturation

  • Analytical Considerations:

    • High-throughput sequencing of both initial libraries and selected pools to quantify enrichment

    • Computational analysis to disentangle binding modes even when epitopes cannot be experimentally isolated

    • Integration of biophysical models to interpret selection results and predict behavior of unobserved variants

For challenging specificity objectives, such as discriminating between highly similar epitopes, researchers should consider implementing combinatorial selection schemes with multiple target combinations. This approach facilitates computational separation of binding contributions and enables the identification of antibodies with customized specificity profiles .

What are the key considerations when analyzing antibody light chains for diagnostic applications?

Antibody light chain analysis has emerged as a powerful diagnostic tool, particularly for plasma cell disorders. Researchers should consider several critical factors:

  • Sample Preparation:

    • For intact light chain analysis, selective immunocapture using heavy chain-specific antibodies provides isotype information

    • For tryptic peptide analysis, careful digestion conditions must be optimized to maintain reproducibility

    • Assessment of post-translational modifications requires specialized approaches, as these can confound clonality assessment3

  • Mass Spectrometry Parameters:

    • MALDI-TOF analysis provides rapid assessment but may miss subtle variants

    • Liquid chromatography coupled with electrospray ionization mass spectrometry (LC-ESI-MS) offers superior resolution for complex samples

    • Careful calibration is essential, as mass shifts can lead to misinterpretation of results3

  • Data Interpretation:

    • Establishment of reference ranges for normal kappa:lambda ratios across different immunoglobulin classes

    • Recognition that sensitivity improvements may detect immune responses previously below detection limits

    • Integration with clinical context and complementary testing (bone marrow assessment, imaging) for comprehensive interpretation3

The technical sensitivity of modern light chain analysis approaches can detect minimal residual disease at levels comparable to or potentially exceeding bone marrow-based assessments, making them valuable tools for monitoring treatment efficacy in multiple myeloma and related disorders3.

What methodological approaches can resolve contradictory results in antibody specificity testing?

When faced with contradictory results in antibody specificity testing, researchers should implement systematic troubleshooting strategies:

  • Multi-platform Validation:

    • Compare results across orthogonal methods (ELISA, surface plasmon resonance, cell-based assays, mass spectrometry)

    • Evaluate binding under varying conditions (pH, ionic strength, temperature) to identify context-dependent interactions

    • Incorporate competitive binding assays to assess relative affinities and potential allosteric effects

  • Epitope Mapping:

    • Implement fine epitope mapping to identify specifically which residues contribute to observed binding differences

    • Utilize mutagenesis studies to confirm the role of specific amino acids in controversial binding interactions

    • Apply hydrogen-deuterium exchange mass spectrometry to characterize conformational aspects of binding

  • Computational Modeling:

    • Employ biophysics-informed models to disentangle multiple binding modes that may explain seemingly contradictory results

    • Identify potential heterogeneity in antibody populations that could account for diverse binding profiles

    • Simulate binding energetics under different conditions to explain context-dependent observations

  • Biological Validation:

    • Assess functional outcomes of binding (signaling, internalization, complement activation)

    • Evaluate binding in increasingly complex environments approaching physiological conditions

    • Confirm specificity in relevant biological samples rather than purified antigens alone

This systematic approach can often resolve apparent contradictions by revealing different binding modes, conformational states, or context-dependent interactions that explain variable experimental outcomes 4.

How might computational design approaches transform antibody engineering beyond current experimental limitations?

Computational approaches are poised to revolutionize antibody engineering by transcending the limitations of traditional experimental methods:

  • Beyond Library Size Constraints:

    • Computational models trained on experimental data can explore sequence space orders of magnitude larger than physical libraries

    • Biophysics-informed models can predict binding properties of antibody sequences never tested experimentally

    • Virtual library screening can identify promising candidates for targeted experimental validation

  • Custom Specificity Profile Design:

    • Energy function optimization enables the design of antibodies with precisely defined cross-reactivity patterns

    • Computational approaches can identify antibody sequences that discriminate between highly similar epitopes

    • Models can predict and mitigate potential off-target interactions before experimental validation

  • Integration with Structural Biology:

    • Machine learning models informed by structural data can predict conformational aspects of antibody-antigen interactions

    • Design of antibodies targeting specific epitope conformations becomes feasible

    • Rational engineering of framework regions to support optimal CDR positioning

Recent research demonstrates the power of these approaches, showing that computational models can successfully disentangle different binding modes associated with chemically similar ligands, even when these cannot be experimentally isolated. Furthermore, these models can generate entirely new antibody sequences with customized specificity profiles that perform as predicted when tested experimentally .

What emerging analytical technologies might enhance our understanding of antibody structure-function relationships?

Several cutting-edge technologies are transforming our ability to characterize antibody structure-function relationships:

  • High-Resolution Mass Spectrometry:

    • Native mass spectrometry enables analysis of intact antibody complexes, revealing stoichiometry and binding dynamics

    • Ion mobility mass spectrometry provides insights into conformational states relevant to function

    • Advances in top-down proteomics allow characterization of antibody variants with single-amino acid resolution3

  • Single-Molecule Techniques:

    • Single-molecule FRET (Förster Resonance Energy Transfer) reveals dynamic conformational changes during antigen binding

    • Atomic force microscopy characterizes mechanical properties of antibody-antigen interactions

    • Single-molecule tracking in cellular environments elucidates the dynamics of antibody-mediated processes

  • Cryo-Electron Microscopy:

    • High-resolution structures of antibody-antigen complexes in near-native states

    • Visualization of conformational ensembles rather than single static structures

    • Insights into the structural basis of polyspecificity and cross-reactivity

  • Integrative Multi-omics Approaches:

    • Combination of structural data with functional genomics and proteomics

    • Systems biology frameworks linking antibody structural features to downstream immune responses

    • Machine learning integration of diverse data types to predict novel structure-function relationships

These technologies are enabling researchers to move beyond static models of antibody function toward dynamic, context-dependent understanding of how structural features determine specific functional outcomes in complex biological environments 34.

How can researchers leverage antibody engineering to address challenges in targeting evolutionarily conserved epitopes?

Targeting evolutionarily conserved epitopes presents unique challenges that require sophisticated engineering approaches:

  • Accessibility Challenges:

    • Conserved epitopes are often buried or masked by variable regions

    • Engineering approaches must focus on unusual binding geometries that can access recessed sites

    • Strategic use of smaller binding domains or non-conventional binding scaffolds may overcome steric limitations

  • Affinity-Specificity Balance:

    • Conserved regions often share structural similarity with host proteins

    • Computational modeling can help identify subtle differences between pathogen and host epitopes

    • Fine-tuning of CDR residues to maximize specificity while maintaining sufficient affinity requires iterative optimization

  • Multi-epitope Targeting Strategies:

    • Design of antibodies that simultaneously engage conserved and variable regions

    • Development of bispecific constructs combining breadth and specificity

    • Engineering of antibodies that recognize conformational signatures unique to pathogens despite sequence conservation

  • Evolutionary Considerations:

    • Analysis of natural antibody repertoires that successfully target conserved epitopes

    • Integration of phylogenetic data to identify natural solutions to similar challenges

    • Prediction of potential escape mutations and pre-emptive design of antibodies resistant to such changes

Recent advances in computational design and biophysics-informed modeling provide powerful tools for addressing these challenges, allowing researchers to design antibodies that recognize subtle differences between highly similar epitopes while maintaining broad coverage of variant sequences 4.

What are the optimal methods for monitoring minimal residual disease in multiple myeloma using antibody detection techniques?

Monitoring minimal residual disease (MRD) in multiple myeloma has evolved significantly with advanced antibody detection techniques. Current optimal approaches include:

  • Mass Spectrometry-Based Methods:

    • The mass fix technique provides superior sensitivity compared to traditional serum protein electrophoresis

    • LC-ESI-MS offers even higher sensitivity, potentially surpassing bone marrow-based methods

    • These techniques can detect M-proteins at levels below 10^-6 (one cancer cell per million normal cells)3

  • Complementary Methodologies:

    • Integration of serum mass spectrometry with bone marrow-based assessments (next-generation sequencing or flow cytometry)

    • The value of serum assays lies particularly in their ability to detect negative results with high confidence

    • Bone marrow techniques provide additional value when positive, creating a powerful complementary approach3

  • Longitudinal Monitoring Strategy:

    • Baseline characterization of the specific M-protein molecular weight and post-translational modification profile

    • Regular monitoring at defined intervals following treatment

    • Quantitative assessment of M-protein levels using internal standards

    • Attention to potential immunoglobulin class switching or light chain escape phenomena

Research has demonstrated that patients who achieve negative results by high-sensitivity mass spectrometry methods show significantly longer progression-free survival compared to those with persistent disease. This approach offers the advantage of less invasive monitoring through serum samples rather than requiring repeated bone marrow aspirations3.

How can researchers distinguish between authentic antibody responses and experimental artifacts in selection experiments?

Distinguishing genuine antibody responses from artifacts in selection experiments requires rigorous quality control measures:

  • Control Strategies:

    • Implementation of parallel selections against irrelevant targets to identify non-specific binders

    • Use of pre-adsorption steps to eliminate binders to matrix components or constant regions

    • Incorporation of isotype-matched control antibodies throughout experimental workflows

  • Biophysical Validation:

    • Confirmation of binding using multiple orthogonal methods (ELISA, SPR, BLI, flow cytometry)

    • Characterization of binding kinetics to distinguish high-affinity specific interactions from low-affinity non-specific binding

    • Assessment of specificity through competitive binding assays with related and unrelated antigens

  • Computational Analysis:

    • Implementation of biophysics-informed models that can identify and account for non-specific binding modes

    • Analysis of enrichment patterns across multiple selection rounds to distinguish consistent enrichment from stochastic fluctuations

    • Examination of sequence features associated with known technical artifacts (expression bias, aggregation propensity)

  • Functional Validation:

    • Confirmation that selected antibodies perform expected biological functions beyond simple binding

    • Correlation of binding properties with functional outcomes

    • Verification in progressively more complex and physiologically relevant systems

Recent research demonstrates the value of computational approaches that incorporate "pseudo-modes" into models to account for biases that may occur during phage production and antibody expression stages, enabling more accurate discrimination between true binding and technical artifacts .

What strategies can optimize antibody research in resource-limited settings?

Conducting sophisticated antibody research in resource-limited settings requires strategic adaptations:

  • Technological Adaptations:

    • Implementation of simplified ELISA formats that require minimal equipment

    • Development of portable, robust analytical systems with reduced infrastructure requirements

    • Utilization of smartphone-based imaging and analysis for quantitative assessments

    • Adoption of ambient-temperature stable reagents to overcome cold chain limitations

  • Collaborative Models:

    • Establishment of hub-and-spoke models where complex analyses are centralized

    • Sample stabilization protocols enabling transportation to central facilities

    • Virtual collaboration platforms for data analysis and interpretation

    • Shared access to computational resources for complex modeling

  • Priority Setting:

    • Focus on high-impact applications most relevant to local health priorities

    • Adaptation of methods to locally prevalent pathogens and conditions

    • Development of point-of-care applications that maximize clinical utility with minimal resource requirements

  • Training and Capacity Building:

    • Implementation of standardized protocols with comprehensive troubleshooting guides

    • Development of training programs focused on robust, reproducible techniques

    • Peer-to-peer knowledge sharing networks to disseminate best practices

    • Training in computational approaches that can maximize the value of limited experimental data 34

By thoughtfully adapting technologies and approaches to local contexts while maintaining scientific rigor, researchers can conduct meaningful antibody research even with significant resource constraints, potentially addressing critical local health needs while contributing to the global scientific knowledge base.

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.