DNER Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Research Applications and Protocols

FITC-conjugated DNER antibodies are pivotal in:

  • Autoimmune Disorder Diagnosis: Detecting anti-Tr/DNER autoantibodies in PCD and Hodgkin’s lymphoma. A recombinant cell-based immunofluorescence assay (RC-IFA) demonstrated 100% sensitivity and specificity for anti-DNER detection .

  • Neuroimmunology: Studying cerebellar degeneration mechanisms via DNER’s interaction with Notch pathways .

  • COPD Research: Profiling DNER-expressing macrophages in lung tissue to investigate IFNγ regulation and emphysema progression .

Example Protocol for Immunofluorescence

  1. Fixation: Treat cells or tissue sections with acetone or formalin.

  2. Incubation: Apply FITC-conjugated DNER antibody (1:300–1:1,200 dilution) for 30 minutes at room temperature .

  3. Detection: Use fluorescence microscopy to visualize FITC emission (green signal) .

Table 1: Select Studies Using FITC-Conjugated Antibodies for DNER Detection

StudyMethodKey OutcomeCitation
Paraneoplastic Cerebellar DegenerationRC-IFA with FITC-labeled anti-human IgGAnti-DNER antibodies identified in 100% of PCD patients (n=38) with Hodgkin’s lymphoma .
COPD PathogenesisFlow cytometry with FITC-anti-IFNγDNER regulates macrophage IFNγ release via Notch1/NF-κB signaling, contributing to emphysema .
Epitope MappingCompetitive inhibition assaysAnti-Tr antibodies bind DNER’s extracellular domain, validated via FITC-based blocking experiments .

Advantages of FITC Conjugation

  • High Sensitivity: FITC’s quantum yield (~90%) ensures low background noise .

  • Multiplex Compatibility: Paired with TRITC or Cyanine dyes for multi-target detection .

  • Stability: Shelf life of 24–48 months under -20°C storage .

Limitations and Considerations

  • Photobleaching: FITC degrades under prolonged light exposure; alternatives like Cyanine 5.5 are preferred for long imaging .

  • Cross-Reactivity: Validate using HEK293 cells transfected with DNER vs. controls (e.g., CDR2/Yo) .

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 your order within 1-3 business days of receiving it. Delivery time may vary depending on your location and chosen shipping method. Please consult your local distributor for specific delivery information.
Synonyms
bet antibody; Brain EGF repeat-containing transmembrane protein antibody; Bret antibody; Delta and Notch-like epidermal growth factor-related receptor antibody; Delta notch like EGF repeat containing transmembrane antibody; Delta/notch like EGF repeat containing antibody; Delta/notch-like EGF-related receptor antibody; Dner antibody; DNER_HUMAN antibody; PRO299 antibody; Transmembrane protein Bet antibody; UNQ26 antibody
Target Names
DNER
Uniprot No.

Target Background

Function
DNER Antibody, FITC conjugated is an activator of the NOTCH1 pathway. It may mediate neuron-glia interaction during astrocytogenesis.
Gene References Into Functions
  1. Quantum dot-based immunofluorescent imaging and quantitative analytical system (QD-IIQAS) has been identified as an easy and accurate method for assessing DNER expression. Research suggests that DNER expression is an independent prognostic factor in prostate cancer. PMID: 29843212
  2. Recent studies have uncovered an unexpected transcriptional repression function of the BET bromodomain, revealing a novel mechanism for TAZ upregulation. PMID: 27717711
  3. Targeting BET proteins for degradation has emerged as a promising therapeutic strategy for the treatment of Triple-negative breast cancers (TNBC). PMID: 28209615
  4. DNER is not a Notch ligand. PMID: 27622512
  5. The structural biology of BET family BDs has been extensively reviewed and discussed, highlighting their applications in major diseases. PMID: 27240990
  6. A study examining a Chinese Han population found that the DNER rs1861612 C to T change and variant T genotype may contribute to type 2 diabetes mellitus (T2DM). PMID: 25300688
  7. Research indicates that DNER is a susceptibility gene for T2DM in American Indians. PMID: 24101674
  8. Studies have revealed that clathrin-independent endocytosis is crucial for the polarized targeting of somatodendritic proteins, including DNER. PMID: 20367751
  9. Inhibition of DNER protein has been shown to enhance adipocyte maturation, partly by reducing cell proliferation through increased CCAAT-Enhancer-Binding Protein-delta expression. PMID: 20070733
  10. DNER is expressed in both developing and mature central nervous systems. PMID: 11950833

Show More

Hide All

Database Links

HGNC: 24456

OMIM: 607299

KEGG: hsa:92737

STRING: 9606.ENSP00000345229

UniGene: Hs.234074

Subcellular Location
Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed in brain, spinal cord and adrenal gland.

Q&A

What is DNER and why is it significant in neurological research?

DNER (Delta/Notch-like epidermal growth factor-related receptor) is a transmembrane protein primarily expressed in brain, spinal cord, and adrenal gland tissues. It functions as an activator of the NOTCH1 pathway and may mediate neuron-glia interaction during astrocytogenesis . DNER has gained significant attention in neurological research due to its role as the target antigen for anti-Tr antibodies, which are associated with paraneoplastic cerebellar degeneration (PCD) and Hodgkin lymphoma . During cerebellar development, DNER interacts with the Notch1 receptor and is involved in the differentiation of Bergmann glia . The receptor's extracellular domain has been identified as the main epitope recognized by anti-Tr antibodies, making it a crucial antigen in autoimmune neurological disorders .

What are the typical applications of FITC-conjugated DNER antibodies in research?

FITC-conjugated DNER antibodies are primarily used in fluorescence-based detection methods including:

  • Flow cytometry (especially intracellular staining) for quantitative analysis of DNER expression in cell populations .

  • Immunofluorescence microscopy for localization studies of DNER in tissue sections and cell cultures.

  • Visualization of DNER distribution in neuronal and glial cells during development and in pathological conditions.

  • Diagnostic testing for anti-DNER autoantibodies in patients suspected of paraneoplastic neurological syndromes.

  • Investigation of Notch signaling pathway components in various experimental models.

These applications leverage the fluorescent properties of FITC conjugation to enable direct visualization without requiring secondary antibody labeling steps, thereby streamlining experimental workflows and potentially reducing background signal .

How does the specificity of DNER antibodies compare to other Notch pathway-related antibodies?

DNER antibodies demonstrate high specificity when properly validated. In standardized recombinant cell-based indirect immunofluorescence assay (RC-IFA) studies, anti-DNER antibodies showed 100% sensitivity (95% CI 92.8%–100%) and 100% specificity (95% CI 98.7%–100%) in detecting DNER-expressing cells without cross-reactivity to control substrates .

Unlike other Notch pathway antibodies such as anti-Notch1, DNER antibodies specifically recognize the Delta/Notch-like epidermal growth factor-related receptor without cross-reacting with other Notch family members or ligands. This specificity is critical for distinguishing DNER-mediated signaling from other Notch pathway components .

Comparative specificity testing has revealed that anti-DNER antibodies do not cross-react with cells expressing other neuronal antigens such as CDR2/Yo and CDR2L, underscoring their specificity for DNER . This high specificity makes DNER antibodies valuable tools for investigating specific roles of DNER in Notch signaling and neuronal-glial interactions.

What is the significance of using recombinant DNER in antibody production and validation?

Recombinant DNER production has revolutionized both antibody development and validation procedures in several key ways:

  • Standardization: Using recombinant DNER fragments (such as those within amino acids 200-350) as immunogens produces antibodies with consistent epitope recognition .

  • Epitope mapping: Recombinant technology has enabled precise identification of the extracellular domain of DNER as the main target of anti-Tr antibodies, clarifying the molecular basis of autoimmune responses .

  • Validation controls: Recombinant DNER-expressing cells provide excellent positive controls for antibody validation, while mock-transfected cells serve as negative controls .

  • Adsorption studies: Preadsorption of sera with recombinant full-length DNER or its extracellular domain selectively abolishes anti-Tr reactivity, confirming antibody specificity .

  • Diagnostic improvements: Recombinant cell-based assays using DNER-expressing HEK293 cells have shown superior performance compared to traditional tissue-based assays, particularly for detecting low-titer antibodies .

The use of recombinant DNER has significantly improved the reliability of anti-DNER antibody detection and characterization, advancing both research applications and clinical diagnostics .

How do different fixation and permeabilization protocols affect FITC-conjugated DNER antibody performance in immunofluorescence studies?

Fixation and permeabilization protocols significantly impact the performance of FITC-conjugated DNER antibodies due to several technical considerations:

Fixation impact:

  • Paraformaldehyde fixation (typically 2-4%) preserves DNER epitopes while maintaining cellular architecture, making it the preferred fixation method for most applications .

  • Over-fixation can mask epitopes in the extracellular domain of DNER, particularly affecting antibodies targeting amino acids 200-350, requiring optimization of fixation duration and concentration.

  • The transmembrane nature of DNER means that improper fixation can alter its conformation and accessibility.

Permeabilization considerations:

  • FITC-conjugated antibodies targeting intracellular domains of DNER require adequate permeabilization (typically with 0.1-0.3% Triton X-100 or 0.1% saponin).

  • For antibodies targeting the extracellular domain, excessive permeabilization may increase background fluorescence and reduce signal-to-noise ratio.

  • Sequential fixation-permeabilization often yields better results than simultaneous protocols for DNER detection.

Protocol optimization:

  • Temperature control during fixation/permeabilization is critical for preserving FITC fluorescence.

  • Antigen retrieval may be necessary for paraffin-embedded tissues but should be optimized to avoid FITC degradation.

  • Buffer composition (particularly pH) can significantly affect both antibody binding and FITC fluorescence intensity.

These considerations necessitate protocol optimization specific to the exact epitope recognized by the DNER antibody and the tissue/cell type under investigation .

What are the experimental considerations when using FITC-conjugated DNER antibodies for detecting paraneoplastic anti-Tr antibodies?

When employing FITC-conjugated DNER antibodies for paraneoplastic anti-Tr antibody detection, researchers should consider several critical experimental factors:

Sample preparation:

  • Serum samples require dilution (typically 1:10 to 1:100) to minimize background without losing sensitivity.

  • Cerebrospinal fluid (CSF) samples may provide higher specificity but require appropriate concentration and handling protocols .

  • Pre-adsorption steps with recombinant DNER extracellular domain can confirm specificity of detected antibodies .

Assay design:

  • Recombinant cell-based indirect immunofluorescence assay (RC-IFA) using HEK293 cells expressing DNER shows superior sensitivity (100%, 95% CI 92.8%–100%) and specificity (100%, 95% CI 98.7%–100%) compared to conventional tissue-based assays .

  • Control substrates including mock-transfected cells and cells expressing other neuronal antigens (e.g., CDR2/Yo) are essential to exclude false positives .

  • Parallel testing of multiple samples from the same patient (serum and CSF) improves diagnostic accuracy.

Result interpretation:

  • The characteristic punctate immunoreactivity pattern must be distinguished from non-specific binding.

  • Low-titer antibodies may be missed by conventional tissue-based assays but detected by RC-IFA with DNER-expressing cells .

  • Correlation with clinical presentation is essential, as anti-Tr/DNER antibodies are strongly associated with PCD and Hodgkin lymphoma .

This methodological approach has been validated in studies involving patients with anti-Tr antibodies, PCD without anti-Tr, Hodgkin lymphoma without neurological symptoms, and appropriate control groups .

How can multiplexed immunofluorescence approaches incorporate FITC-conjugated DNER antibodies for advanced neurological research?

Multiplexed immunofluorescence incorporating FITC-conjugated DNER antibodies enables sophisticated analyses of neurological tissues and pathways through several advanced approaches:

Spectral compatibility considerations:

  • FITC emission (peak ~520 nm) must be spectrally separated from other fluorophores in multiplexed panels.

  • Compatible fluorophores include Cy3, Cy5, or Alexa Fluor 647 for other target proteins.

  • Use of quantum dots with narrow emission spectra can further expand multiplexing capacity while including FITC-conjugated DNER antibodies.

Co-localization studies:

  • DNER co-localization with Notch1 receptors can be assessed using FITC-DNER antibodies paired with alternative fluorophore-conjugated Notch1 antibodies .

  • Neuronal-glial interactions can be visualized by combining DNER detection with astrocytic markers.

  • Dendritic targeting of DNER can be analyzed relative to synaptic proteins using appropriate fluorophore combinations.

Technical optimization for multiplexing:

  • Sequential staining protocols may be necessary to avoid cross-reactivity.

  • Tyramide signal amplification can enhance FITC signals for detecting low-abundance epitopes while allowing multiple rounds of staining.

  • Automated multispectral imaging platforms with spectral unmixing algorithms help resolve overlapping signals in complex multiplexed panels.

Research applications:

  • Developmental neurobiology studies tracking DNER expression alongside differentiation markers.

  • Pathological investigations examining relationship between DNER and immune cell infiltration in PCD.

  • Mechanistic studies of Notch pathway activation in various neurological disorders.

This multiplexed approach supports comprehensive spatial and contextual analysis of DNER expression and function within complex neural circuits and pathological states .

What are the methodological differences between using DNER antibodies for research versus diagnostic applications?

The application of DNER antibodies differs significantly between research and diagnostic contexts across several methodological dimensions:

ParameterResearch ApplicationsDiagnostic Applications
Antibody validationExtensive characterization including Western blot, immunoprecipitation, and knockout controlsStandardized validation against reference panels of positive and negative samples
Sample typesDiverse (cell lines, animal tissues, human samples)Primarily human serum or CSF samples
Detection systemsVarious (direct fluorescence, amplification methods)Standardized protocols with defined cutoff values
ControlsExperimental controls specific to research questionMandatory positive, negative, and specificity controls
InterpretationFocused on relative expression, localization, or functional outcomesBinary (positive/negative) with potential titer determination
Conjugation preferencesVarious fluorophores selected based on experimental designStandardized conjugates (FITC or equivalent) for consistent results
Performance metricsEmphasis on reproducibility and specificityCritical focus on clinical sensitivity (100%, 95% CI 92.8%–100%) and specificity (100%, 95% CI 98.7%–100%)

Research applications typically employ DNER antibodies to investigate fundamental biological questions regarding DNER function, whereas diagnostic applications focus on reliable detection of anti-DNER autoantibodies in patient samples. The recombinant cell-based indirect immunofluorescence assay (RC-IFA) has emerged as superior to traditional tissue-based assays for diagnostic purposes, particularly for samples with low-titer antibodies . This methodological distinction underscores the importance of selecting appropriate protocols based on the intended application.

What controls should be included when validating a new lot of FITC-conjugated DNER antibody?

Comprehensive validation of new FITC-conjugated DNER antibody lots requires a systematic approach with multiple control elements:

Positive controls:

  • Recombinant HEK293 cells expressing full-length DNER to confirm specific binding .

  • Tissues or cell lines with known DNER expression (brain, spinal cord, or adrenal gland samples) .

  • Previously validated antibody lot tested in parallel for direct comparison.

Negative controls:

  • Mock-transfected HEK293 cells to assess non-specific binding .

  • Cells expressing related proteins (CDR2/Yo, CDR2L) to evaluate cross-reactivity .

  • Tissues known to lack DNER expression as specificity controls.

  • Isotype control antibodies conjugated to FITC to identify non-specific binding.

Specificity verification:

  • Preadsorption with recombinant DNER extracellular domain to confirm epitope specificity .

  • Western blot or immunoprecipitation to verify recognition of the expected 78 kDa protein .

  • Peptide competition assays with the immunogenic peptide (amino acids 200-350) .

  • Signal reduction in DNER-knockdown or knockout models.

Fluorophore integrity assessment:

  • Spectral analysis to confirm proper FITC excitation/emission characteristics.

  • Photobleaching tests to ensure stability during imaging.

  • Assessment of fluorescence signal-to-noise ratio compared to previous lots.

This comprehensive validation approach ensures both the specificity of the antibody for DNER and the functionality of the FITC conjugation, minimizing experimental variability and enhancing reproducibility .

How does sample preparation affect the detection of DNER using FITC-conjugated antibodies in different tissue types?

Sample preparation significantly influences DNER detection across different tissue types when using FITC-conjugated antibodies:

Brain and cerebellar tissues:

  • Fresh frozen sections preserve native DNER epitopes but require careful handling to maintain tissue architecture.

  • Paraformaldehyde fixation (4%) followed by sucrose cryoprotection preserves the characteristic punctate staining pattern of DNER in cerebellar tissues .

  • Antigen retrieval (citrate buffer, pH 6.0) is often necessary for formalin-fixed paraffin-embedded (FFPE) cerebellar sections but must be optimized to prevent FITC degradation .

Cell cultures and cell lines:

  • For HEK293 cells expressing recombinant DNER, mild fixation (2% paraformaldehyde, 10 minutes) is typically sufficient .

  • Neuronal cultures require careful optimization of permeabilization to access intracellular domains while preserving membrane-associated DNER.

  • Suspension cells for flow cytometry require specific fixation/permeabilization protocols compatible with flow analysis .

Lymphatic tissues (for Hodgkin lymphoma studies):

  • FFPE lymph node tissues require more aggressive antigen retrieval protocols to unmask DNER epitopes.

  • Multiplexed approaches for simultaneous detection of DNER and lymphoma markers require sequential staining protocols.

  • Higher antibody concentrations may be necessary for lymphatic tissues compared to neural tissues.

Technical considerations across tissue types:

  • Autofluorescence: Brain tissues exhibit higher autofluorescence in the FITC spectrum, requiring specific quenching steps.

  • Background reduction: Blocking protocols must be optimized per tissue type (5% normal serum, 1% BSA recommended for neural tissues).

  • Signal amplification: Tyramide signal amplification may be necessary for tissues with low DNER expression.

  • pH sensitivity: FITC fluorescence is pH-sensitive, requiring careful buffer optimization (ideally pH 7.4-8.0).

These tissue-specific considerations ensure optimal detection of DNER while minimizing artifacts and false signals across experimental systems .

What troubleshooting approaches can address weak or non-specific signals when using FITC-conjugated DNER antibodies?

When encountering signal issues with FITC-conjugated DNER antibodies, systematic troubleshooting approaches can resolve common technical challenges:

For weak signal:

  • Antibody concentration optimization:

    • Titrate antibody concentration using positive control samples

    • Consider longer incubation times (4°C overnight instead of 1-2 hours at room temperature)

  • Sample processing adjustment:

    • Optimize fixation time to prevent epitope masking

    • Evaluate alternative antigen retrieval methods (heat vs. enzymatic)

    • Increase permeabilization for intracellular epitopes

  • Signal enhancement strategies:

    • Implement tyramide signal amplification system

    • Use anti-FITC antibodies conjugated to brighter fluorophores

    • Adjust imaging parameters (exposure time, gain) while maintaining control comparisons

For non-specific signal:

  • Background reduction:

    • Implement more stringent blocking (5-10% normal serum from the same species as secondary antibody)

    • Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions

    • Include 0.1-0.3M NaCl in wash buffers to reduce ionic interactions

  • Cross-reactivity elimination:

    • Pre-adsorb antibody with recombinant DNER to confirm specificity

    • Use appropriate controls (isotype controls, secondary-only controls)

    • Compare staining pattern with known DNER distribution

  • Fluorescence troubleshooting:

    • Minimize sample exposure to light to prevent photobleaching

    • Use fresh mounting medium with anti-fade properties

    • Counter potential pH issues by maintaining buffer pH at 7.4-8.0

Validation approaches:

  • Pattern recognition:

    • Verify that staining shows the characteristic punctate pattern in cerebellar Purkinje cells

    • Compare with alternative anti-DNER antibodies from different sources

  • Alternative detection methods:

    • Confirm DNER presence using non-FITC conjugated antibodies

    • Validate with Western blot or RT-PCR in parallel

This structured troubleshooting approach systematically addresses technical issues while maintaining experimental rigor and validity .

How can researchers optimize detection protocols when using FITC-conjugated DNER antibodies for flow cytometry applications?

Optimizing flow cytometry protocols for FITC-conjugated DNER antibodies requires systematic technical adjustments to maximize signal quality and quantitative accuracy:

Sample preparation optimization:

  • Cell dissociation considerations:

    • Enzymatic dissociation must preserve DNER epitopes (mild trypsinization or non-enzymatic dissociation preferred)

    • Single-cell suspensions are critical for accurate analysis

    • Viable cell selection using appropriate exclusion dyes (e.g., 7-AAD)

  • Fixation/permeabilization strategy:

    • For intracellular DNER detection: 0.1% saponin or commercial permeabilization buffers designed for flow cytometry

    • For surface epitopes: Avoid harsh detergents that may disrupt membrane integrity

    • Temperature control during fixation (4°C preferred to minimize internalization)

Staining protocol refinement:

  • Antibody titration:

    • Systematic titration to determine optimal concentration (typically 1-10 μg/mL)

    • Evaluation using signal-to-noise ratio rather than absolute signal intensity

    • Incubation time optimization (30-60 minutes at 4°C typically sufficient)

  • Buffer optimization:

    • Use of protein-containing buffers (1-2% BSA) to reduce non-specific binding

    • Addition of 0.1% sodium azide to prevent internalization during staining

    • pH control to maximize FITC fluorescence (pH 7.4-8.0)

Instrument setup and analysis:

  • Cytometer configuration:

    • Proper voltage settings for FITC channel determined using unstained and single-stained controls

    • Compensation with other fluorophores when performing multicolor analysis

    • Regular QC with standardized beads to ensure consistent detection

  • Gating strategy:

    • Initial gating on viable single cells

    • Comparison with isotype-matched FITC-conjugated control antibodies

    • When analyzing patient samples for anti-DNER autoantibodies, include HEK293 DNER-transfected cells as positive controls

Validation approaches:

  • Controls for each experiment:

    • Unstained cells for autofluorescence assessment

    • Isotype-matched FITC-conjugated antibody controls

    • Positive controls (DNER-expressing cells) and negative controls (mock-transfected cells)

  • Functional validation:

    • Correlation of DNER expression with functional parameters

    • Blocking studies with recombinant DNER to confirm specificity

These optimizations ensure reliable quantitative assessment of DNER expression or anti-DNER autoantibodies in various experimental and clinical applications .

How do DNER antibody detection methods compare in sensitivity and specificity for diagnosing paraneoplastic cerebellar degeneration?

Different DNER antibody detection methods demonstrate varying performance characteristics for paraneoplastic cerebellar degeneration (PCD) diagnosis:

Detection MethodSensitivitySpecificityKey AdvantagesLimitations
Recombinant Cell-Based IFA (RC-IFA)100% (95% CI 92.8%–100%) 100% (95% CI 98.7%–100%) Superior detection of low-titer antibodies; Standardized approach Requires specialized cell culture facilities
Commercial Tissue-Based IFALower (missed 4 samples with low-titer antibodies) HighWidely availableLess sensitive for low-titer antibodies; Requires expert interpretation
Conventional ImmunohistochemistryVariableVariableHistorical standard methodLabor-intensive; Requires epitope blocking confirmation
Western BlotLimitedHighConfirms molecular weightPoor for conformational epitopes; Low sensitivity
ELISANot well established for DNERNot well established for DNERHigh-throughput potentialMay miss conformational epitopes

The recombinant cell-based indirect immunofluorescence assay (RC-IFA) using HEK293 cells expressing DNER has emerged as the gold standard, combining maximal sensitivity and specificity . This method outperforms traditional tissue-based approaches particularly for samples with low-titer antibodies, which might be missed by commercial tissue-based assays .

In a comprehensive evaluation of 38 anti-Tr positive samples and 201 control samples (including patients with PCD without anti-Tr, Hodgkin lymphoma without neurological symptoms, rheumatic diseases, and healthy donors), RC-IFA demonstrated perfect sensitivity and specificity . This makes it the preferred method for diagnosing anti-Tr/DNER-associated paraneoplastic cerebellar degeneration, with important implications for timely tumor screening and treatment initiation .

What are the methodological challenges in studying DNER expression in Hodgkin lymphoma tissues using immunofluorescence techniques?

Investigating DNER expression in Hodgkin lymphoma presents several distinct methodological challenges for immunofluorescence-based approaches:

Tissue architecture considerations:

  • Hodgkin lymphoma's characteristic disrupted architecture with sparse Reed-Sternberg cells complicates identification of DNER-expressing cells.

  • The inflammatory microenvironment creates high background when using fluorescence-based detection.

  • FFPE lymphoma samples often require aggressive antigen retrieval that can affect FITC stability.

Technical optimization requirements:

  • Signal-to-noise ratio enhancement:

    • Tyramide signal amplification may be necessary to detect low-level DNER expression.

    • Multiplex panels with CD30/CD15 (Hodgkin markers) require careful fluorophore selection to avoid spectral overlap with FITC.

    • Autofluorescence quenching (using Sudan Black B or commercial reagents) is critical for lymph node tissues.

  • Antibody validation challenges:

    • Limited availability of positive controls with known DNER expression in lymphoid tissues.

    • Need for parallel evaluation with multiple anti-DNER antibodies targeting different epitopes.

    • Critical importance of isotype controls due to non-specific binding from tissue-resident immunoglobulins.

Research design considerations:

  • Sample selection:

    • Requirement for properly staged and classified Hodgkin lymphoma samples.

    • Need for paired samples (tumor and patient serum) to correlate DNER expression with autoantibody development.

    • Inclusion of appropriate controls (reactive lymph nodes, non-Hodgkin lymphomas).

  • Analytical approaches:

    • Quantification challenges due to heterogeneous expression patterns.

    • Need for digital image analysis with machine learning algorithms to identify rare DNER-positive cells.

    • Requirement for co-localization analysis with lineage markers to identify specific DNER-expressing cell populations.

These methodological challenges necessitate careful experimental design and technical optimization to accurately characterize DNER expression in Hodgkin lymphoma and its relationship to the development of paraneoplastic neurological syndromes .

How can researchers design experiments to investigate the functional consequences of anti-DNER antibodies in neuronal tissues?

Designing experiments to elucidate anti-DNER antibody functional effects requires sophisticated approaches across multiple model systems:

In vitro neuronal culture models:

  • Primary cerebellar cultures:

    • Application of purified patient-derived anti-DNER antibodies to cerebellar cultures

    • Live cell imaging with calcium indicators to assess acute electrophysiological effects

    • Quantification of dendritic complexity and synaptic markers after chronic antibody exposure

    • Co-culture with glial cells to assess DNER-mediated neuron-glia interaction disruption

  • Cerebellar slice cultures:

    • Organotypic cerebellar slices treated with anti-DNER antibodies at varying concentrations

    • Electrophysiological recording of Purkinje cell activity and circuit function

    • Immunofluorescence analysis of DNER internalization and degradation following antibody binding

    • Assessment of Notch pathway activation using reporter systems

In vivo approaches:

  • Passive transfer models:

    • Intrathecal injection of purified anti-DNER antibodies into rodent models

    • Cerebellar-specific behavioral testing (rotarod, beam walking, gait analysis)

    • Terminal histopathological analysis for complement deposition and inflammatory markers

    • Electrophysiological studies of cerebellar circuit function

  • Active immunization models:

    • Immunization with recombinant DNER to generate in vivo autoimmune responses

    • Correlation of antibody titers with cerebellar function

    • Therapeutic intervention studies targeting specific immune components

Molecular mechanism investigations:

  • Antibody engineering approaches:

    • Comparison of effects between intact IgG, F(ab')2, and Fab fragments to distinguish Fc-dependent and Fc-independent effects

    • Creation of monoclonal antibodies targeting specific DNER epitopes to map functional domains

    • Humanized mouse models expressing human Fc receptors for translational relevance

  • Signaling pathway analysis:

    • Quantification of Notch pathway activation using reporter assays

    • Phosphoproteomic analysis of cerebellar tissues following anti-DNER exposure

    • Transcriptomic analysis to identify disrupted gene networks

These experimental approaches provide complementary insights into the pathophysiological mechanisms through which anti-DNER antibodies contribute to cerebellar dysfunction, potentially revealing therapeutic targets for paraneoplastic cerebellar degeneration .

What methodological approaches can distinguish anti-DNER antibodies from other autoantibodies associated with paraneoplastic neurological syndromes?

Methodological differentiation of anti-DNER antibodies from other paraneoplastic autoantibodies requires multi-layered analytical approaches:

Immunofluorescence pattern analysis:

  • Tissue-based screening:

    • Anti-DNER/Tr produces a characteristic punctate staining of cerebellar molecular layer and punctate dendritic labeling of Purkinje cells .

    • This pattern differs from anti-Yo (Purkinje cell cytoplasm), anti-Hu (neuronal nuclei), and anti-Ri (neuronal nuclei) patterns.

    • Expert interpretation of these patterns provides initial differentiation but requires confirmation .

  • Recombinant cell-based assays:

    • Parallel testing using a panel of HEK293 cells expressing different recombinant antigens:

      • DNER-expressing cells for anti-Tr

      • CDR2/Yo-expressing cells for anti-Yo

      • HuD-expressing cells for anti-Hu

    • This approach showed 100% sensitivity and specificity for distinguishing anti-DNER from other antibodies .

Molecular confirmation techniques:

  • Epitope-specific assays:

    • Preadsorption studies using recombinant DNER extracellular domain selectively abolish anti-Tr reactivity .

    • Competition assays with defined peptide fragments can map epitope specificity.

    • Indirect immunoprecipitation followed by mass spectrometry for novel autoantibody characterization.

  • Multiplex approaches:

    • Luminex-based multiplex assays using different recombinant antigens coupled to distinguishable beads.

    • Line/dot blot assays with multiple recombinant antigens for parallel screening.

    • Protein microarrays for high-throughput autoantibody profiling.

Clinical and functional correlation:

  • Clinical phenotype assessment:

    • Anti-DNER/Tr: Strong association with PCD and Hodgkin lymphoma (particularly in younger patients) .

    • Other antibodies have distinct clinical and tumor associations (anti-Yo with gynecological/breast cancers, anti-Hu with small cell lung cancer).

    • Age and sex distribution analysis helps refine diagnostic suspicion.

  • Antibody characteristics:

    • Isotype and subclass determination (most anti-DNER antibodies are IgG1).

    • Intrathecal synthesis assessment (CSF/serum antibody index).

    • Epitope spreading analysis in longitudinal samples.

This comprehensive approach ensures accurate distinction between anti-DNER and other paraneoplastic autoantibodies, which is critical for appropriate tumor screening and treatment planning .

How might single-cell techniques advance our understanding of DNER function using fluorescently-labeled antibodies?

Single-cell technologies coupled with fluorescently-labeled DNER antibodies open new frontiers for understanding DNER biology at unprecedented resolution:

Single-cell transcriptomics integration:

  • FITC-conjugated DNER antibodies can be used for fluorescence-activated cell sorting (FACS) to isolate DNER-expressing populations for subsequent single-cell RNA sequencing.

  • This approach enables correlation between DNER protein expression levels and comprehensive transcriptomic profiles.

  • Trajectory analysis of DNER-expressing cells during development or in disease states can reveal dynamic regulatory networks.

  • Identification of cell-type specific co-expression patterns may uncover novel DNER interaction partners.

Spatial transcriptomics applications:

  • In situ sequencing or spatial transcriptomics platforms combined with DNER immunofluorescence create spatially-resolved molecular maps.

  • These techniques preserve tissue architecture while revealing transcriptional states of DNER-expressing and neighboring cells.

  • Analysis of spatial relationships between DNER-expressing cells and other cell types may uncover paracrine signaling mechanisms.

  • Three-dimensional reconstruction of DNER distribution across brain regions at single-cell resolution.

Mass cytometry approaches:

  • Metal-conjugated DNER antibodies enable high-parameter mass cytometry (CyTOF) analysis.

  • Simultaneous measurement of DNER with dozens of other proteins provides detailed cellular phenotyping.

  • Unsupervised clustering algorithms can identify previously unrecognized DNER-expressing cell populations.

  • Single-cell proteomic analysis of signaling pathway activation states in DNER-positive cells.

Live-cell dynamics:

  • FITC-conjugated Fab fragments of DNER antibodies enable live imaging of DNER trafficking and turnover.

  • Single-molecule tracking microscopy can reveal DNER diffusion dynamics and clustering behaviors.

  • Optogenetic manipulation combined with DNER labeling allows direct assessment of activity-dependent regulation.

  • CRISPR-based lineage tracing of DNER-expressing cells through development.

These emerging single-cell approaches promise to transform our understanding of DNER function in neurodevelopment, Notch signaling, and neurological disorders at unprecedented resolution .

What novel imaging techniques might enhance the utility of FITC-conjugated DNER antibodies in neurological research?

Advanced imaging technologies are poised to revolutionize DNER visualization and functional analysis in neurological research:

Super-resolution microscopy applications:

  • Stimulated Emission Depletion (STED) microscopy can resolve DNER nanoscale organization beyond diffraction limits (~20nm resolution).

  • Single Molecule Localization Microscopy (PALM/STORM) enables precise mapping of DNER distribution at molecular scale.

  • Structured Illumination Microscopy (SIM) provides ~100nm resolution suitable for DNER localization within dendritic spines.

  • These approaches can reveal previously undetectable patterns of DNER clustering and co-localization with Notch pathway components .

Three-dimensional imaging innovations:

  • Light-sheet microscopy allows rapid imaging of DNER distribution throughout intact cleared brain tissues.

  • Two-photon excitation microscopy enables deep tissue imaging of FITC-labeled DNER in living tissues with reduced phototoxicity.

  • Expansion microscopy physically enlarges specimens for enhanced visualization of DNER subcellular localization.

  • Volume electron microscopy combined with correlative light microscopy links DNER immunofluorescence to ultrastructural context.

Functional imaging integration:

  • Genetically-encoded calcium indicators combined with DNER immunolabeling correlate activity with receptor expression.

  • Optogenetic stimulation coupled with DNER visualization reveals activity-dependent receptor dynamics.

  • FRET-based approaches using FITC-conjugated DNER antibodies and acceptor-labeled binding partners detect molecular interactions.

  • Label-free imaging techniques (Raman microscopy, second harmonic generation) provide complementary structural information.

Multiplexed imaging platforms:

  • Cyclic immunofluorescence (CycIF) allows sequential imaging of dozens of markers in the same tissue section alongside DNER.

  • Mass spectrometry imaging (MIBI, IMC) enables highly multiplexed analysis with metal-conjugated DNER antibodies.

  • DNA-barcoded antibody methods (CODEX) facilitate highly multiplexed imaging of DNER in complex cellular environments.

  • AI-assisted image analysis for automated quantification of complex DNER expression patterns.

These emerging technologies will provide unprecedented insights into DNER biology in development, disease, and potential therapeutic applications .

How might engineered DNER antibody fragments advance both research and therapeutic applications?

Engineered DNER antibody fragments offer transformative potential across both research and therapeutic domains:

Research applications:

  • Improved imaging probes:

    • Single-chain variable fragments (scFvs) conjugated to FITC provide smaller probes for improved tissue penetration

    • Site-specific conjugation technologies for optimal fluorophore positioning

    • Bispecific antibody fragments for simultaneous targeting of DNER and interaction partners

    • pH-sensitive fluorescent conjugates to track DNER internalization and trafficking

  • Functional manipulation tools:

    • Photoswitchable antibody fragments for spatiotemporal control of DNER function

    • PROTAC-conjugated fragments for targeted DNER degradation

    • Split-fluorescent protein complementation for visualization of DNER dimerization

    • Intrabodies for tracking and manipulating DNER in living cells

Therapeutic potential:

  • Diagnostic applications:

    • Engineered fragments with optimized binding to DNER for improved diagnostic sensitivity

    • Bispecific formats simultaneously capturing DNER and secondary markers

    • Radiotracer development for PET imaging of DNER expression in neurological disorders

    • Affinity-tuned variants for distinguishing pathogenic vs. physiological anti-DNER antibodies

  • Therapeutic development:

    • Blocking antibody fragments to prevent pathogenic autoantibody binding to DNER

    • Fc-engineered antibodies with reduced complement activation for neuroprotection

    • Blood-brain barrier penetrating formats (e.g., transferrin receptor-DNER bispecifics)

    • Antibody-drug conjugates for targeted therapy of DNER-expressing Hodgkin lymphoma cells

Technical innovations required:

  • Production advances:

    • Phage display libraries for selection of high-affinity DNER-binding fragments

    • Yeast surface display for affinity maturation and stability engineering

    • Site-specific conjugation technologies for optimal FITC positioning

    • Humanization strategies for therapeutic development

  • Validation requirements:

    • Comprehensive epitope mapping to design non-competing diagnostic and therapeutic antibodies

    • Functional screening assays to identify fragments that modulate DNER-Notch signaling

    • Animal models to evaluate blood-brain barrier penetration and target engagement

These engineered antibody approaches represent the frontier of DNER research, potentially transforming both basic neuroscience and clinical applications for paraneoplastic neurological disorders .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.