DOGL2 Antibody

Shipped with Ice Packs
In Stock

Description

Absence of Direct References

None of the nine provided sources mention "DOGL2 Antibody" or variants of this term. The search results focus on antibodies targeting well-characterized canine antigens, including:

  • CD20 (e.g., monoclonal antibodies like 4E1-7-B for lymphoma treatment )

  • CD18 (e.g., YFC 118.12 for immune disorders )

  • Desmoglein-2 (associated with arrhythmogenic right ventricular cardiomyopathy in Boxer dogs )

  • Canine parvovirus antibodies (e.g., IgM/IgG therapies )

Terminology Issues

  • Misspelling or Mislabeling: "DOGL2" may represent a typographical error or nonstandard abbreviation. For example, "DOG" is sometimes used informally for "dog," but no established canine antibody uses this prefix.

  • Hypothetical Target: The term might refer to an uncharacterized or proprietary antigen not yet published in peer-reviewed studies.

Research Gaps

  • Antibody development in veterinary medicine often prioritizes targets with direct clinical relevance (e.g., CD20 for lymphoma, desmoglein-2 for cardiomyopathy). Novel antibodies like "DOGL2" may lack published validation.

Related Antibody Development in Canine Research

While "DOGL2" remains unidentified, current efforts in canine antibody research include:

Table 1: Key Canine Antibodies in Development/Use

TargetAntibody NameApplicationSource Catalog (Example)
CD204E1-7-B, 1E4-BB-cell lymphoma therapy
CD18YFC 118.12Leukocyte adhesion deficiency
PD-1/PD-L1JC053, JC071Immunotherapy for cancers
Desmoglein-2(Polyclonal)ARVC diagnosis in Boxer dogs

Recommendations for Further Inquiry

  • Verify Terminology: Confirm the correct spelling or nomenclature of "DOGL2" with the original source.

  • Explore Synonyms: Investigate whether "DOGL2" corresponds to an alternative name for a known antigen (e.g., LY6 family proteins, which include some "L2" designations).

  • Consult Proprietary Databases: Patent repositories or biotech company pipelines may list unpublished antibodies.

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
DOGL2 antibody; At4g18680 antibody; F28A21.90Protein DOG1-like 2 antibody
Target Names
DOGL2
Uniprot No.

Q&A

What is the relationship between desmoglein-2 antibodies and cardiac conditions in dogs?

Desmoglein-2 (DSG2) is a desmosomal protein that has been implicated in cardiac diseases, particularly arrhythmogenic right ventricular cardiomyopathy (ARVC). While autoantibodies against desmoglein-2 have been associated with ARVC in humans, research in canines has yielded different results. Studies have detected anti-desmoglein-2 antibodies in all dogs tested, regardless of breed or cardiac disease status . This includes Boxers with ARVC, healthy Boxers, Doberman Pinschers with dilated cardiomyopathy, various small breeds with myxomatous mitral valve disease, and healthy control dogs. This suggests that, unlike in humans, the mere presence of these antibodies is not specifically diagnostic for ARVC in canines .

What methods are used to detect desmoglein-2 antibodies in canine samples?

Detection of desmoglein-2 antibodies in canine samples typically employs immunoblot analysis. The methodology involves:

  • Validation using commercially available anti-desmoglein-2 antibodies on recombinant desmoglein-2

  • Denaturation, reduction, and separation of proteins by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE)

  • Identification of protein bands at the expected molecular weight (~115 kDa)

  • Semi-quantitative measurement via densitometry

  • Normalization to globulin concentrations in serum for comparative analysis

This technique allows for both qualitative detection and relative quantification of antibody levels across different sample groups.

How should researchers interpret the presence of antibodies in control and disease groups?

When antibodies are detected in both control and disease groups, interpretation requires careful consideration. In the case of anti-desmoglein-2 antibodies, their presence in all dogs regardless of disease status suggests they may represent naturally occurring antibodies rather than disease-specific markers . Researchers should:

  • Compare antibody concentrations (not just presence/absence) between groups

  • Normalize data to account for baseline differences in globulin levels

  • Consider correlations with clinical parameters and disease severity

  • Evaluate potential functional differences in antibodies between groups

  • Investigate potential epitope differences that standard detection methods might miss

The finding that antibody expression did not correlate significantly with age, body weight, or disease status emphasizes the importance of functional studies rather than mere detection.

What immunological assays are most appropriate for evaluating antibody functionality in canine models?

When evaluating antibody functionality in canine models, researchers should consider multiple complementary assays:

  • Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC): This assay measures the ability of antibodies to induce target cell lysis through effector cells. Typically, target cells (e.g., CLBL-1/luc cells) are incubated with the antibody of interest and IL-2-stimulated peripheral blood lymphocytes at defined effector-to-target ratios (e.g., 20:1). Cytotoxicity can be calculated from luminometer data using the equation: percent specific lysis = 100 × (spontaneous death RLU – test RLU)/(spontaneous death RLU – maximal killing RLU) .

  • Complement-Dependent Cytotoxicity (CDC): This measures the ability of antibodies to activate complement cascade leading to cell lysis. Different antibody isotypes may show variable CDC activity, as observed with canine IgG-B and IgG-C subclasses .

  • Cell Proliferation Assays: These evaluate the direct effect of antibodies on cell growth, with or without crosslinking. For example, cells can be incubated with antibodies for 72 hours, followed by addition of CCK-8 solution and measurement of absorbance at 450 nm .

These functional assays provide more valuable insights than mere binding studies, particularly when developing therapeutic antibodies or investigating pathogenic mechanisms.

How can researchers optimize Western blotting protocols for canine antibody detection?

Optimizing Western blotting for canine antibody detection requires careful attention to several methodological details:

  • Sample Preparation:

    • Collect and store cell lines at -80°C until use

    • Perform proper pre-clearing of cell lysates with protein A/G agarose (10 μl) for 1 hour at 4°C with rotation

    • Ensure consistent protein loading across samples

  • Immunoprecipitation Protocol:

    • Mix pre-cleared supernatant with 1 μg of each antibody (premixed with 10 μl of protein A/G agarose) at 4°C overnight

    • Wash immunoprecipitates thoroughly (three times with PBS)

  • Detection System:

    • Select appropriate primary antibodies (e.g., anti-Flag, anti-actin mouse monoclonal antibody)

    • Use matched HRP-conjugated secondary antibodies (anti-rat, anti-mouse)

    • Visualize using chemiluminescence reagent and sensitive imaging systems like Luminescent Image Analyzer

  • Controls and Validation:

    • Include positive controls with known reactivity

    • Validate molecular weight markers for target proteins (e.g., ~115 kDa for desmoglein-2)

    • Use increasing loading amounts of recombinant protein as positive controls

Optimized protocols are essential for reliable detection and semi-quantitative analysis, particularly when comparing antibody levels between different disease states.

What are the limitations of current methods for correlating antibody expression with clinical disease severity?

Current methods for correlating antibody expression with clinical disease severity face several significant limitations:

  • Quantification Challenges:

    • Densitometry provides only semi-quantitative measurements

    • Variable background signals can affect interpretation

    • Normalization approaches (e.g., to globulin levels) may introduce biases

  • Population Heterogeneity:

    • Age differences between study groups can confound results

    • Breed-specific variations in antibody expression

    • Comorbidities may influence antibody levels independent of the primary disease

  • Functional Relevance:

    • Presence of antibodies does not necessarily indicate pathogenicity

    • Similar antibody levels may have different functional impacts between individuals

    • Multiple epitopes may exist for the same protein, with varying clinical significance

  • Temporal Considerations:

    • Single time-point measurements miss dynamic changes in antibody levels

    • Disease progression may correlate better with antibody fluctuations than absolute levels

    • Treatment effects on antibody expression are often not accounted for

Research addressing these limitations should incorporate longitudinal sampling, functional assays, and multivariate analysis approaches to better understand the complex relationship between antibody expression and disease manifestation.

How do different antibody isotypes and subclasses affect functional outcomes in canine immunology research?

Different antibody isotypes and subclasses significantly impact functional outcomes in canine immunology research, as evidenced by several observations:

  • ADCC Activity Variations:

    • Studies show that chimeric antibodies with canine IgG-B chains demonstrate higher ADCC activity compared to those with IgG-C chains

    • Some antibodies (e.g., 1E4-B) may show no ADCC activity despite having other effector functions like antibody-dependent cellular phagocytosis (ADCP)

  • CDC Activity Differences:

    • Canine IgG-B and IgG-C may have differential CDC activity

    • CDC activity does not always correlate with C1q binding ability

    • Experimental conditions (incubation time, temperature) can significantly impact observed CDC activity

  • Direct Effects on Cell Proliferation:

    • Chimerization can alter antibody characteristics, potentially enhancing ADCC and CDC while reducing direct suppressive effects on cell proliferation

    • Crosslinking of antibodies can enhance their functional effects, as demonstrated with anti-dog IgG crosslinking

  • Glycosylation Impact:

    • Defucosylation can provide additive ADCC activity to antibodies

    • Post-translational modifications significantly influence antibody effector functions

Understanding these differences is crucial when developing therapeutic antibodies or when using antibodies as research tools, as the choice of isotype or subclass can dramatically affect experimental outcomes.

What approaches can distinguish naturally occurring antibodies from disease-associated autoantibodies?

Distinguishing naturally occurring antibodies from disease-associated autoantibodies requires sophisticated analytical approaches:

  • Epitope Mapping:

    • Disease-associated autoantibodies may target specific epitopes not recognized by naturally occurring antibodies

    • Peptide arrays or competition assays can help identify epitope differences

  • Affinity Analysis:

    • Disease-associated autoantibodies often display higher affinity

    • Binding kinetics studies using flow cytometry with serially diluted antibodies can determine Kd values

    • Non-linear regression analysis (e.g., Michaelis-Menten curve fit) can quantify binding differences

  • Functional Assays:

    • Pathogenic autoantibodies typically demonstrate functional effects in vitro

    • Comparing ADCC, CDC, and direct cellular effects between patient and control antibodies

    • Cell-based assays measuring disruption of specific cellular functions

  • Longitudinal Analysis:

    • Monitoring changes in antibody levels over disease course

    • Correlation with disease flares and remissions

    • Response to immunosuppressive therapy

  • Isotype and Subclass Distribution:

    • Disease-associated autoantibodies may show skewed isotype distributions

    • Analysis of light chain usage (κ vs λ) may reveal differences

This multi-faceted approach can help researchers distinguish clinically relevant autoantibodies from naturally occurring antibodies that may have different biological significance.

What control groups should be included when studying antibodies in canine disease models?

Comprehensive control group selection is critical when studying antibodies in canine disease models. Based on research methodologies:

  • Disease-Specific Controls:

    • Include dogs with different cardiac diseases (e.g., dilated cardiomyopathy, myxomatous mitral valve disease) when studying a specific condition like ARVC

    • This helps distinguish disease-specific from general cardiac disease markers

  • Breed-Specific Controls:

    • Include healthy dogs of the same breed as affected dogs (e.g., healthy Boxers when studying Boxers with ARVC)

    • This controls for breed-specific variations in antibody expression

  • Diverse Healthy Controls:

    • Include healthy dogs of various breeds

    • This establishes normal baseline variation across the canine population

  • Age and Size-Matched Controls:

    • Match control groups for age and body weight

    • This is important as these factors may influence antibody expression independently of disease

  • Positive Technical Controls:

    • Include known positive samples or recombinant proteins at varying concentrations

    • This validates detection methods and establishes assay sensitivity

What statistical approaches are most appropriate for analyzing antibody expression data in comparative studies?

Analyzing antibody expression data in comparative studies requires careful statistical consideration:

  • Normalization Strategies:

    • Normalize raw densitometry data to globulin concentrations to account for baseline differences in antibody levels

    • Compare both raw and normalized data to ensure robust findings

  • Group Comparisons:

    • Use ANOVA with appropriate post-hoc tests (e.g., Tukey's) when comparing multiple groups

    • Report specific p-values for each comparison rather than just significance thresholds

  • Correlation Analysis:

    • Examine correlations between antibody levels and:

      • Clinical parameters (e.g., arrhythmia burden)

      • Demographic factors (age, weight)

      • Biochemical markers

    • Report correlation coefficients (r) with confidence intervals

  • Multivariate Analysis:

    • Consider multiple variables simultaneously to identify independent predictors

    • Account for confounding factors like age, breed, and comorbidities

  • Sample Size Considerations:

    • Conduct power analysis to determine appropriate sample sizes

    • Report confidence intervals to indicate precision of estimates

    • Acknowledge limitations when sample sizes are small

These approaches enhance the robustness of findings and reduce the risk of spurious associations, particularly important when studying complex biological phenomena like antibody expression.

How can researchers validate novel antibodies for specificity and sensitivity in canine studies?

Validating novel antibodies for canine studies requires a systematic approach to ensure specificity and sensitivity:

  • Target Verification:

    • Express recombinant target proteins with tags (e.g., Flag tag)

    • Confirm antibody binding to the tagged protein at the expected molecular weight

    • Perform immunoprecipitation followed by mass spectrometry

  • Cross-Reactivity Testing:

    • Test against cells expressing versus not expressing the target

    • Examine binding to closely related proteins to assess specificity

    • Use knockout/knockdown systems when available

  • Flow Cytometry Validation:

    • Perform titration experiments with serially diluted antibodies

    • Calculate binding affinity (Kd value) using Nonlinear Regression Michaelis–Menten curve fit

    • Compare binding profiles across multiple cell types

  • Immunoblotting Confirmation:

    • Compare binding patterns between positive and negative control samples

    • Verify expected molecular weight of detected proteins

    • Test antibody specificity under reducing and non-reducing conditions

  • Functional Validation:

    • Assess functional activity through ADCC, CDC, or direct effects assays

    • Compare with established antibodies when available

    • Evaluate epitope binding through competition assays

Thorough validation ensures that experimental findings reflect true biological phenomena rather than technical artifacts, particularly important when developing novel research or therapeutic antibodies.

What are promising approaches for enhancing antibody effector functions in canine therapeutic applications?

Several promising approaches exist for enhancing antibody effector functions in canine therapeutic applications:

  • Glycoengineering:

    • Defucosylation has been demonstrated to provide tenfold stronger ADCC activity compared to regular antibodies

    • This approach has shown efficacy in enhancing peripheral B cell depletion in healthy beagles

  • Isotype Selection and Engineering:

    • Selecting optimal canine IgG subclasses (e.g., IgG-B showing stronger ADCC and CDC activity compared to IgG-C)

    • Engineering chimeric antibodies that combine desirable properties of different isotypes

  • Crosslinking Strategies:

    • Enhancing direct effects of antibodies through crosslinking approaches

    • Developing bispecific antibodies that can engage multiple targets simultaneously

  • Combination Therapies:

    • Using antibodies in combination with conventional therapies (e.g., CHOP chemotherapy for lymphoma)

    • Developing antibody-drug conjugates for targeted delivery

  • Epitope Optimization:

    • Identifying and targeting epitopes that maximize therapeutic effects

    • Engineering antibodies with optimized binding to functional domains of target proteins

These approaches hold significant promise for improving outcomes in canine patients with conditions ranging from cancer to autoimmune diseases, potentially providing more effective and targeted therapeutic options.

What challenges remain in translating human antibody research findings to canine applications?

Translating human antibody research to canine applications faces several significant challenges:

  • Immunological Differences:

    • Canine Fc receptors and complement systems differ from humans

    • Effector mechanisms like ADCC and CDC may have different relative importance between species

  • Target Homology Considerations:

    • Despite conservation, key epitopes may differ between human and canine proteins

    • Amino acid differences in target molecules can affect antibody binding and function

  • Antibody Structure Variations:

    • Canine IgG subclasses (A, B, C, D) differ from human IgG1-4

    • Glycosylation patterns and their impact on function may vary between species

  • Clinical Translation Hurdles:

    • Limited availability of canine-specific reagents for research

    • Smaller market size affecting commercial development

    • Regulatory pathways less established for veterinary biologics

  • Contrasting Disease Mechanisms:

    • Disease pathophysiology may differ between humans and dogs

    • Unexpected findings, such as anti-desmoglein-2 antibodies being present in all dogs regardless of disease status, highlight species-specific differences

Understanding and addressing these challenges is crucial for successful translation of antibody-based therapies from human medicine to veterinary applications, requiring careful species-specific validation and optimization.

How might novel antibody detection methods improve diagnostic capabilities in veterinary medicine?

Novel antibody detection methods have significant potential to enhance diagnostic capabilities in veterinary medicine:

  • Multiplex Assay Platforms:

    • Simultaneous detection of multiple antibodies from limited sample volumes

    • Integration of autoantibody panels for comprehensive disease profiling

    • Improved sensitivity compared to traditional Western blotting approaches

  • Single B-Cell Isolation and Antibody Cloning:

    • Direct isolation of disease-specific antibodies from affected animals

    • Characterization of antibody repertoires in health and disease

    • Identification of specific pathogenic clones

  • High-Throughput Epitope Mapping:

    • Peptide arrays to identify specific epitopes targeted in disease

    • Distinguishing pathogenic from non-pathogenic antibody responses

    • Development of epitope-specific diagnostics

  • Point-of-Care Testing:

    • Rapid lateral flow assays for antibody detection in clinical settings

    • Microfluidics-based detection systems for quantitative results

    • Smartphone-integrated reading systems for field use

  • Functional Antibody Assays:

    • Moving beyond presence/absence to measure pathogenic potential

    • Cell-based reporter systems detecting antibody-mediated effects

    • Correlation of functional metrics with clinical outcomes

These advances could transform veterinary diagnostics from simple detection to comprehensive characterization of antibody responses, enabling more precise diagnosis, prognosis, and therapeutic monitoring.

What are the optimal conditions for flow cytometry analysis of canine antibodies?

Optimal flow cytometry analysis of canine antibodies requires careful attention to technical details:

  • Sample Preparation:

    • Collect cells and wash with flow cytometry buffer (PBS with 2% FBS and 0.1% NaN₃)

    • Use consistent cell numbers (e.g., 2 × 10⁵ cells) for staining

    • Maintain samples at 4°C during processing to preserve antibody binding

  • Antibody Staining Protocol:

    • Primary staining: Use appropriate antibody dilutions determined by titration

    • Secondary detection: Select compatible labeled antibodies (PE-labeled anti-rat IgG, Dylight 649-labeled anti-rat IgG, or Alexa 647-labeled anti-dog IgG)

    • Include proper isotype controls (e.g., rat IgG₂ₐ for rat-derived antibodies)

  • Subclass Determination:

    • For novel antibodies, determine subclass using biotin-labeled secondary antibodies specific for different isotypes (e.g., anti-rat IgG-κ, anti-rat IgG-λ, anti-rat IgG₁, anti-rat IgG₂ₐ, anti-rat IgG₂ₑ)

    • Follow with streptavidin-PE for detection

  • Binding Affinity Determination:

    • Stain cells with serially diluted antibodies

    • Determine Kd value using Nonlinear Regression Michaelis–Menten curve fit

    • Use specialized software (e.g., JMP14.0) for analysis

  • Data Analysis:

    • Analyze results using dedicated software (e.g., FlowJo v.10)

    • Apply consistent gating strategies across samples

    • Use fluorescence minus one (FMO) controls for accurate gate setting

These optimized conditions ensure reliable and reproducible flow cytometry data for canine antibody characterization, essential for both research and diagnostic applications.

What considerations are important when developing in vivo models to test antibody efficacy?

Developing in vivo models to test antibody efficacy requires careful consideration of several key factors:

  • Model Selection:

    • Choose between xenotransplant models in immunodeficient mice or canine models

    • Consider that mouse models allow controlled experimentation but may not fully recapitulate canine immune responses

    • Healthy beagles provide relevant immunological context but introduce greater biological variability

  • Dosing Regimens:

    • Determine optimal dosing based on pharmacokinetic studies

    • Consider that even single administration can induce considerable effects (e.g., peripheral B cell depletion)

    • Establish clear endpoints for efficacy assessment

  • Cell Line Selection for Xenografts:

    • Use well-characterized cell lines (e.g., CLBL-1 for canine B cell lymphoma)

    • Consider engineering cells to express luciferase for in vivo tracking

    • Validate target expression in the cell line before in vivo studies

  • Monitoring Approaches:

    • Implement comprehensive monitoring of peripheral blood cell populations

    • Track tumor growth in xenograft models

    • Assess both target depletion and downstream physiological effects

  • Ethical and Regulatory Considerations:

    • Adhere to animal welfare guidelines

    • Use the minimum number of animals required for statistical significance

    • Consider pilot studies to refine protocols and reduce animal use

Careful attention to these considerations enhances the translational value of in vivo studies while maintaining ethical standards, ultimately improving the predictive value for clinical applications.

What are the key methodological differences between studying natural autoantibodies versus therapeutic monoclonal antibodies?

Studying natural autoantibodies versus therapeutic monoclonal antibodies involves distinct methodological approaches:

ParameterNatural AutoantibodiesTherapeutic Monoclonal Antibodies
SourcePolyclonal mixture from serum/plasmaSingle clone, recombinantly produced
Detection MethodsWestern blotting with target protein Flow cytometry, ELISA, immunoprecipitation
QuantificationSemi-quantitative densitometry; normalization to globulin levels Absolute quantification; standard curves with known concentrations
Functional AssaysFocus on pathogenic mechanisms (e.g., signaling disruption)Focus on therapeutic effects (ADCC, CDC, direct effects)
Specificity TestingAbsorption studies with target antigensExtensive cross-reactivity testing against related targets
Heterogeneity AnalysisEpitope mapping of polyclonal responseSingle defined epitope with affinity measurement
In Vivo StudiesNatural disease modelsXenograft models, dose-finding studies
ControlsHealthy individuals, different disease groups Isotype-matched non-binding antibodies

Understanding these methodological differences is essential for researchers designing studies on either autoantibodies in natural disease processes or therapeutic monoclonal antibodies for clinical applications. Each approach requires specific technical considerations to generate valid and clinically relevant data.

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.