CER26L Antibody

Shipped with Ice Packs
In Stock

Description

Biological Characteristics of CDR2L Antibodies

CDR2L antibodies belong to the IgG class and target a 35 kDa neuronal protein expressed in cerebellar Purkinje cells . These antibodies exhibit:

  • High avidity binding when co-occurring with CDR2 antibodies

  • Low avidity binding when occurring in isolation

  • Epitope specificity distinct from CDR2, despite partial sequence homology

Key structural features align with typical IgG antibodies :

FeatureDescription
Heavy ChainVH domains with somatic hypermutation
Light ChainVL domains (κ or λ)
Antigen-Binding SiteFormed by complementary-determining regions (CDRs) of VH and VL domains

Clinical Associations

CDR2L antibodies show strong links to paraneoplastic cerebellar degeneration (PCD), particularly in ovarian cancer patients :

Table 1. Antibody Prevalence in 36 Dual-Positive Cases

Cancer TypePatientsPCD DiagnosisPurkinje Cell Staining
Ovarian1817Positive
Breast43Positive
Uterine/Fallopian55Positive
Colon11Positive

Key findings:

  • 27/36 (75%) patients with dual CDR2L/CDR2 antibodies had confirmed PCD

  • Isolated CDR2L antibodies (7 cases) showed no PCD association

  • Antibody avidity strongly correlates with neurological symptoms

Diagnostic Performance

Comparative analysis of detection methods:

Table 2. Assay Sensitivity for CDR2L Antibodies

MethodDual Antibodies (n=36)Isolated CDR2L (n=7)
Immunoprecipitation100%100%
Immunofluorescence86%0%
Line Blot83%0%

Critical limitations:

  • Low-avidity antibodies fail standard immunohistochemical detection

  • Transfected cell assays required for isolated CDR2L antibody confirmation

Pathogenic Mechanism

Proposed model of CDR2L antibody activity:

  1. Molecular mimicry between tumor antigens and cerebellar proteins

  2. Complement activation via Fcγ receptor binding

  3. Direct cytotoxicity through Purkinje cell membrane disruption

Notably, co-existing CDR2 antibodies may synergistically enhance blood-brain barrier penetration and target engagement .

Therapeutic Implications

While no targeted therapies exist, clinical management strategies include:

  1. Immunosuppression: IVIG or plasma exchange for acute symptom control

  2. Tumor-directed therapy: Ovarian cancer resection in 18/27 PCD cases improved neurological outcomes

  3. Monitoring: Serial antibody titers correlate with treatment response

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CER26L antibody; At3g23840 antibody; F14O13.3Protein ECERIFERUM 26-like antibody; CER26-like antibody; CER2-like protein 2 antibody; CER2-like2 antibody
Target Names
CER26L
Uniprot No.

Target Background

Function
CER26L is involved in the biosynthesis of the epicuticular wax. It plays a role in very-long-chain fatty acid (VLCFA) biosynthesis and is essential for VLCFA elongation in leaves. Despite its classification as a BAHD acyltransferase based on sequence homology, CER26L does not appear to share the catalytic mechanism of other members of the BAHD family.
Gene References Into Functions
  1. Research indicates that CER2-LIKE proteins, in conjunction with condensing enzymes, are crucial for fertilization. [CER2-LIKE2] PMID: 25596184
Database Links

KEGG: ath:AT3G23840

STRING: 3702.AT3G23840.1

UniGene: At.37641

Protein Families
Plant acyltransferase family
Tissue Specificity
Highly expressed in flowers. Expressed in leaves.

Q&A

What is the fundamental mechanism of action for CER26L Antibody?

The mechanism of action for CER26L Antibody, similar to other therapeutic antibodies, involves specific binding to target proteins. Antibodies typically function by recognizing and binding to specific parts of proteins called spike proteins that act as anchor points for cellular interactions. By blocking these spike proteins, antibodies prevent interaction with cell receptors and thus inhibit associated biological processes. The protective effect occurs through this blocking mechanism, preventing the target protein from attaching to and affecting cells in the body . This fundamental binding process forms the basis for both diagnostic applications and therapeutic interventions in research settings.

How should I design preliminary validation experiments for CER26L Antibody?

When designing preliminary validation experiments for CER26L Antibody, follow these methodological steps:

  • Define your variables carefully - list both independent variables (antibody concentration, incubation time) and dependent variables (binding affinity, specificity)

  • Write a specific, testable hypothesis about CER26L Antibody's effect on your target system

  • Design experimental treatments with appropriate controls:

    • Negative controls (isotype-matched irrelevant antibody, such as anti-GFP)

    • Positive controls (known effective antibody against your target)

  • Assign subjects to experimental groups using either between-subjects or within-subjects design

  • Plan precise measurement methods for your dependent variable

What techniques are most effective for detecting CER26L Antibody binding in tissue samples?

The most effective techniques for detecting CER26L Antibody binding in tissue samples include immunohistochemistry, which provides detailed spatial information about binding patterns. When implementing this approach, surgical specimens should be carefully prepared and evaluated for expression of the target protein. As seen with similar research on CD26 expression in mesothelioma tissues, immunohistochemistry can effectively distinguish between binding patterns in pathological versus normal tissues .

Additional effective detection techniques include:

TechniqueResolutionSample TypeKey Advantages
ImmunohistochemistryCellular/subcellularFixed tissue sectionsSpatial distribution data
Flow cytometryCellularCell suspensionsQuantitative binding assessment
Western blottingProteinTissue lysatesSize verification of target protein
Antibody microarraysProteinMultiple samplesHigh-throughput screening

When selecting techniques, consider the specific research question and whether spatial information or quantitative binding metrics are more valuable for your experimental goals .

How can I assess potential cross-reactivity of CER26L Antibody with related protein families?

Assessing cross-reactivity of CER26L Antibody requires systematic evaluation using complementary methodologies. Begin with computational approaches comparing epitope sequences against protein databases to identify potential cross-reactive targets. Follow with experimental validation using antibody microarray technologies, which enable pairwise combinatorial analysis of protein co-expression .

To rigorously assess cross-reactivity:

  • Design a comprehensive panel of potential cross-reactive proteins

  • Implement a microarray approach with:

    • Your CER26L Antibody as the detection probe

    • Immobilized target proteins and potential cross-reactive proteins

    • Appropriate positive controls (known target proteins)

    • Negative controls (unrelated proteins like GFP)

  • Evaluate signal intensities quantitatively, setting detection thresholds as signal from negative control capture plus 2 standard deviations

  • Validate findings with orthogonal methods such as surface plasmon resonance or competitive binding assays

When interpreting results, remember that background noise can be significant and may explain why some visibly detectable signals fall below statistical thresholds .

What are the optimal experimental conditions for evaluating CER26L Antibody-mediated cell lysis?

The optimal experimental conditions for evaluating antibody-mediated cell lysis, including for CER26L Antibody, should be designed to distinguish between direct cytotoxic effects and immune-mediated cytotoxicity. Based on similar studies with therapeutic antibodies like humanized anti-CD26 monoclonal antibody, the following experimental approach is recommended:

  • Establish baseline cytotoxicity by measuring:

    • Direct antibody effects via cell viability assays in pure cell cultures

    • Antibody-dependent cell-mediated cytotoxicity (ADCC) using co-cultures with immune effector cells

  • Isolate mechanisms by determining:

    • Direct anti-tumor effects through assessment of cellular signaling pathways

    • Evaluation of key regulatory proteins (such as p27kip1 accumulation seen with anti-CD26 mAb)

    • ADCC mechanisms through selective depletion of effector cells

  • Optimize key experimental parameters:

    • Effector-to-target cell ratios (typically ranging from 5:1 to 50:1)

    • Antibody concentrations (dose-response curves)

    • Incubation times (4-72 hours depending on mechanism being evaluated)

These structured experiments will allow for comprehensive characterization of both direct and immune-mediated cytotoxic effects of CER26L Antibody in your experimental system .

How does hybrid immunity affect experimental outcomes when studying CER26L Antibody efficacy?

Hybrid immunity - the combined effect of infection and vaccination - introduces important variables that must be controlled when studying antibody efficacy. When designing experiments to evaluate CER26L Antibody in the context of hybrid immunity, researchers should consider:

  • Subject selection strategies:

    • Categorize subjects based on exposure history (naïve, infection-only, vaccination-only, hybrid)

    • Match groups for demographic factors and time since exposure

    • Consider sequential sampling from the same subjects when possible

  • Analytical approaches:

    • Develop technology similar to Ig-Seq to provide detailed analysis of antibody responses

    • Isolate and characterize antibody sequences and binding properties

    • Compare neutralization patterns across different subject categories

  • Experimental readouts:

    • Measure not only binding affinity but also functional neutralization

    • Assess cross-reactivity against variant forms of targets

    • Evaluate neutralization breadth against related protein families

The study of hybrid immunity in antibody research has revealed that some broadly neutralizing antibodies arise specifically in response to multiple antigenic exposures, providing important context for CER26L Antibody efficacy evaluation .

What control conditions are essential when evaluating CER26L Antibody specificity in vitro?

When evaluating CER26L Antibody specificity in vitro, implement these essential control conditions:

  • Negative binding controls:

    • Isotype-matched irrelevant antibody (e.g., anti-GFP antibody)

    • Target-negative cell lines or tissues

    • Competitive binding with known ligands

  • Positive binding controls:

    • Validated antibodies against the same target

    • Recombinant target protein at known concentrations

    • Target-overexpressing cellular systems

  • Technical controls:

    • Signal calibration standards

    • Background fluorescence measurements

    • Non-specific binding determinants through pre-blocking experiments

Statistical interpretation should include setting detection thresholds based on negative control captures plus 2 standard deviations, as demonstrated in antibody microarray approaches . Remember that significant background noise may occur, explaining why some visible signals might fall below statistical thresholds in quantitative analysis .

How should I design experiments to determine optimal CER26L Antibody concentration for therapeutic applications?

Designing experiments to determine optimal CER26L Antibody concentration requires careful consideration of both efficacy and potential toxicity. Follow this methodology:

  • In vitro dose optimization:

    • Conduct dose-response experiments across a logarithmic concentration range

    • Measure both target engagement (binding) and functional outcomes

    • Determine EC50 (half-maximal effective concentration) values

    • Assess potential off-target effects at higher concentrations

  • In vivo pilot studies:

    • Establish baseline pharmacokinetics with different dosing regimens

    • Monitor antibody half-life and tissue distribution

    • Evaluate preliminary efficacy signals

    • Assess safety parameters and toxicity thresholds

  • Comparative analysis:

    • Create dose-response tables comparing multiple parameters:

Dose LevelTarget EngagementFunctional ResponseTissue PenetrationSide Effects
LowX% bindingY% efficacyLimitedMinimal
MediumX% bindingY% efficacyModerateModerate
HighX% bindingY% efficacyExtensiveSignificant

This structured approach, similar to that used in evaluating therapeutic antibodies like anti-CD26 mAb, provides a comprehensive dataset for determining the optimal therapeutic window .

What experimental design is most appropriate for evaluating CER26L Antibody effects in xenograft models?

For evaluating CER26L Antibody effects in xenograft models, implement a rigorous experimental design that accounts for biological variability while providing clear efficacy signals:

  • Study design framework:

    • Use a randomized block design with stratification based on initial tumor volume

    • Include parallel groups: vehicle control, isotype antibody control, and CER26L Antibody treatment

    • Power analysis should determine sample size (typically 8-12 animals per group)

    • Include both male and female animals to assess sex-specific responses

  • Treatment protocol optimization:

    • Begin treatment when tumors reach a standardized volume (typically 50-100 mm³)

    • Establish multiple dosing regimens (e.g., weekly, biweekly)

    • Consider combination treatments with standard-of-care interventions

  • Comprehensive endpoint analysis:

    • Primary endpoints: tumor volume over time and survival

    • Secondary endpoints: target engagement in tumor tissue, immune infiltration

    • Exploratory endpoints: biomarker modulation, resistance mechanisms

This approach parallels successful xenograft studies with therapeutic antibodies like humanized anti-CD26 mAb, which demonstrated significant tumor growth inhibition and enhanced survival in preclinical models .

How should I analyze conflicting results between in vitro and in vivo CER26L Antibody studies?

When confronted with discrepancies between in vitro and in vivo CER26L Antibody studies, implement this methodical approach to analysis:

  • Systematic comparison of experimental conditions:

    • Analyze antibody concentration differences between systems

    • Evaluate matrix effects present in vivo but absent in vitro

    • Assess pharmacokinetic/pharmacodynamic differences

    • Consider immune system contributions present only in vivo models

  • Mechanistic investigation:

    • Determine if differences stem from direct antibody effects versus immune-mediated mechanisms

    • Evaluate potential metabolic modifications of the antibody in vivo

    • Consider target accessibility differences between systems

  • Resolution strategies:

    • Develop intermediate model systems (ex vivo tissue cultures, organoids)

    • Implement matched experimental conditions where possible

    • Design new experiments specifically addressing hypothesized sources of discrepancy

When analyzing such discrepancies, remember that in vivo systems often reveal complex mechanisms not apparent in vitro, such as antibody-dependent cell-mediated cytotoxicity that may contribute significantly to therapeutic efficacy but remain undetectable in pure cell culture systems .

What statistical approaches are most appropriate for analyzing CER26L Antibody binding specificity data?

For analyzing CER26L Antibody binding specificity data, employ these statistical approaches:

  • Primary statistical methods:

    • Set detection thresholds as signal from negative control capture plus 2 standard deviations

    • Implement multivariate analysis to control for background noise

    • Use paired statistical tests when comparing binding across different targets

    • Apply false discovery rate correction for multiple comparisons

  • Visualization and interpretation techniques:

    • Generate heat maps of binding across target panels

    • Implement hierarchical clustering to identify binding patterns

    • Create receiver operating characteristic (ROC) curves to optimize specificity/sensitivity tradeoffs

  • Advanced analytical considerations:

    • Account for antibody concentration effects through standardized binding ratios

    • Normalize signals to account for protein abundance differences

    • Implement machine learning approaches for pattern recognition in complex datasets

These methods parallel approaches used in antibody microarray analysis, where background noise can be significant and careful statistical treatment is required to distinguish true signals from artifacts .

How can I effectively interpret CER26L Antibody data from multiple cell line models?

Effective interpretation of CER26L Antibody data across multiple cell line models requires structured analytical approaches that account for biological variability while identifying consistent patterns:

  • Systematic cross-model comparison:

    • Normalize data within each cell line model before comparison

    • Calculate fold changes relative to appropriate controls in each system

    • Identify consistent effects across models versus cell-specific responses

  • Advanced analytical framework:

    • Implement hierarchical analysis that groups cell lines by characteristics:

      • Tissue of origin

      • Target protein expression levels

      • Genetic background

      • Morphological features

  • Integrated data visualization:

    • Create multi-parameter tables showing key metrics across cell lines:

Cell LineTarget ExpressionBinding AffinityFunctional EffectResponse Duration
Line AHighEC50 = x nM80% inhibition48 hours
Line BMediumEC50 = y nM40% inhibition24 hours
Line CLowEC50 = z nM10% inhibition12 hours

This approach facilitates identification of correlations between target expression and functional outcomes, similar to comparisons seen in studies of antibody effects across different cancer cell lines .

What are the most common causes of false positives in CER26L Antibody binding assays?

  • Technical sources of false positives:

    • Non-specific binding to Fc receptors on target cells

    • Cross-reactivity with structurally similar epitopes

    • Matrix effects from complex biological samples

    • Inappropriate negative control selection

  • Methodological mitigation strategies:

    • Implement Fc blocking reagents in assay buffers

    • Validate results with multiple detection methods

    • Include isotype-matched negative controls (e.g., anti-GFP antibodies)

    • Set detection thresholds based on negative control signal plus 2 standard deviations

  • Analytical approaches to distinguish true signals:

    • Compare binding patterns across multiple related and unrelated targets

    • Implement competitive binding assays with known ligands

    • Validate binding with knockout/knockdown systems

When interpreting binding data, significant background noise can explain why some visibly apparent signals may fall below statistical detection thresholds, necessitating careful analytical approaches .

How can I optimize sample preparation to improve detection sensitivity for CER26L Antibody targets?

Optimizing sample preparation is critical for maximizing detection sensitivity in antibody research. Follow these methodological approaches:

  • Tissue sample optimization:

    • Evaluate multiple fixation protocols to preserve epitope accessibility

    • Implement antigen retrieval techniques appropriate for your target

    • Optimize blocking conditions to minimize background while preserving specific signals

    • Consider fresh-frozen versus fixed samples based on epitope sensitivity

  • Cell preparation considerations:

    • Standardize cell harvesting methods to maintain surface protein integrity

    • Optimize permeabilization protocols for intracellular targets

    • Implement gentle washing techniques to prevent target loss

  • Protein extraction refinements:

    • Select detergents based on target protein characteristics

    • Incorporate protease and phosphatase inhibitors to preserve native states

    • Standardize protein quantification to ensure consistent loading

These optimization approaches can significantly improve signal-to-noise ratios in detection systems, as demonstrated in studies using antibody microarrays for protein detection where sample preparation critically impacts assay performance .

What strategies can address epitope masking when evaluating CER26L Antibody in complex biological samples?

Epitope masking presents a significant challenge when evaluating antibody binding in complex biological samples. Implement these strategies to address this challenge:

  • Sample preparation modifications:

    • Evaluate multiple buffer conditions varying in ionic strength and pH

    • Test different detergent types and concentrations to expose masked epitopes

    • Implement gentle denaturation methods to increase epitope accessibility

    • Consider enzymatic treatments to remove interfering glycosylation

  • Analytical approaches:

    • Compare native versus denaturing conditions to identify masking effects

    • Implement epitope mapping to identify accessible binding regions

    • Develop competition assays with smaller probe molecules

  • Advanced technical solutions:

    • Consider alternative antibody formats (e.g., single-domain antibodies)

    • Implement multiple antibodies targeting different epitopes on the same protein

    • Develop sequential immunoprecipitation approaches to first remove masking proteins

These methodologies parallel approaches used in antibody microarray technologies, where detection of both outer membrane and inner protein components requires careful consideration of accessibility and sample preparation conditions .

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.