CCL15 (chemokine CC-2, leukotactin-1, MIP-1δ, or MIP-5) is a macrophage inflammatory protein that binds CCR1 and CCR3 receptors to induce chemotaxis, immune cell activation, and inflammatory responses . Antibodies targeting CCL15 are critical for studying its role in diseases such as cancer, autoimmune disorders, and viral infections.
Format: CCL15 antibodies are typically IgG isotypes (polyclonal or monoclonal) .
Target Epitopes: Recognize specific regions of CCL15, such as residues Gln22-Ile113 (MAB363) or Ser46-Ile113 (MAB3631) .
Glycosylation: CCL15 antibodies may exhibit glycosylation patterns affecting binding affinity, similar to Fc region glycosylation in general antibody structures .
CCL15 antibodies neutralize chemokine activity by:
Inhibiting downstream signaling pathways (e.g., PLC, PKC, NF-κB) .
Reducing immune cell migration (e.g., macrophages, T cells) .
Autoimmune Disorders: Neutralizing CCL15 antibodies improve clinical outcomes in demyelinating diseases by suppressing T cell trafficking .
Cancer: CCL15 overexpression correlates with tumor metastasis; antibodies may inhibit microenvironment recruitment of protumoral macrophages .
KEGG: ath:AT4G13410
STRING: 3702.AT4G13410.1
CSLA15 Antibody has been validated for several standard immunological techniques. The primary applications include Western blotting (reducing and native conditions), immunohistochemistry (with various fixation methods), and enzyme-linked immunosorbent assays (ELISA). When designing experiments, consider that each application requires specific optimization protocols. For Western blotting, CSLA15 has been validated with cell lysates, tissue lysates, and immunogen-overexpressed lysates . For immunohistochemistry applications, multiple fixation methods have been validated including acetone, ethanol, formalin/paraformaldehyde, and methanol fixation approaches .
Proper experimental controls are critical for antibody-based research. For CSLA15 Antibody work, implement these methodological controls:
Blank control: Wells coated with capture antibody and blocked, but without sample or detector antibodies. This controls for plate variation in ELISA applications and should yield values approaching zero .
Zero concentration control: Include all buffers and reagents without the target antigen, establishing true background levels .
Non-specific binding control: Block wells as usual, then add blocking/wash buffer instead of reagents, followed by labeled detector antibody and substrate. This assesses the detector antibody's contribution to the signal .
Knockout validation: When available, use knockout samples to confirm specificity, particularly for Western blot (WBKO) and immunohistochemistry (IHKO) .
When studying antibody responses longitudinally, timing is crucial. Based on research with similar antibodies, detectable responses typically emerge within 12-14 days post-onset of symptoms (POS) . In longitudinal studies, IgG responses may persist longer, while IgM and IgA responses tend to peak between 20-30 days POS and then decline rapidly, approaching baseline after 60 days . When designing longitudinal studies using CSLA15, collect samples at multiple timepoints (early, peak, and late phase) to accurately capture the complete antibody response dynamics.
For multiplex cytometry using CSLA15 Antibody, several optimization steps are essential. Mass cytometry studies require well-established laboratory protocols and careful panel design . When incorporating CSLA15 into cytometry panels:
Titrate the antibody across several concentrations (typically 1:50, 1:100, 1:200, 1:400, 1:800) to determine optimal signal-to-noise ratio
Validate specificity with appropriate positive and negative controls
Test for potential spectral overlap with other antibodies in your panel
Confirm that fixation and permeabilization methods maintain epitope recognition
For cellular targets of CSLA15, consider enrichment protocols to increase detection sensitivity of rare populations, particularly when working with heterogeneous samples like peripheral blood mononuclear cells.
Single-cell screening methodologies have revolutionized antibody research. For CSLA15 applications at the single-cell level, consider the CelliGO droplet microfluidics system, which combines high-throughput screening for IgG activity using fluorescence-based in-droplet single-cell bioassays with sequencing of paired antibody V genes via in-droplet single-cell barcoded reverse transcription . This approach allows for:
Characterization of antibody binding to both soluble and membrane-bound antigens
Analysis of repertoire diversity
Assessment of clonal expansion and somatic hypermutation
Generation of recombinant antibodies from identified sequences
Such approaches yield approximately 100-1,000 IgG sequences per immunized subject, providing comprehensive data for antibody characterization studies .
CSLA15 Antibody can be employed in various ELISA formats, each with distinct advantages for specific research questions:
Direct ELISA is simpler but has disadvantages, particularly that enzyme reactions must be stopped at predetermined timepoints to distinguish between high and low concentration samples . Sandwich ELISA offers higher sensitivity and specificity through the use of capture and detector antibodies, making it the preferred format for complex biological samples.
When designing experiments to investigate T cell differentiation using CSLA15 Antibody, consider these methodological principles:
Isolation and stimulation: Begin with isolation of naïve CD4+ T cells and stimulate with anti-CD3 and anti-CD28 to activate them. Assess protein levels at different time points post-stimulation (24h, 48h, 72h) .
Polarizing conditions: Culture cells under various polarizing conditions (Th0, Th1, Th2, Th17, Treg) and assess how different cytokine environments affect your target pathway. Analysis of neddylation states may provide insights into protein activation .
Subset analysis: Include comprehensive phenotyping of T cell subsets (CD44+CD4+, effector vs. memory populations) and assessment of associated cytokine production (particularly IL-4, IL-5, IL-9) .
In vivo models: Consider using models of allergic asthma or other inflammatory conditions to assess functional outcomes of pathway manipulation. House dust mite (HDM) exposure models provide physiologically relevant insights into T cell function in vivo .
When troubleshooting CSLA15 Antibody in immunohistochemistry applications, these methodological considerations can help minimize false results:
Sources of False Positives:
Inadequate blocking (especially in tissues with high endogenous biotin or peroxidase activity)
Cross-reactivity with similar epitopes
Overfixation leading to nonspecific binding
Excessive antibody concentration
Inappropriate secondary antibody selection
Sources of False Negatives:
Overfixation masking the epitope
Insufficient antigen retrieval
Antibody concentration too low
Sample degradation
To validate results, compare acrylic/plastic embedding with paraffin embedding, as different preservation methods can significantly affect epitope accessibility and antibody binding characteristics .
To quantify neutralization capacity:
Pseudotype neutralization assay: Establish a dose-response curve with serially diluted antibody preparations to determine the inhibitory dilution 50% (ID50) .
Correlation analysis: Correlate neutralization values with binding data (OD values) from ELISA against relevant antigens. Strong correlations (r² values) between ID50 and OD values indicate reliable quantification .
Temporal considerations: Account for the time-dependent nature of neutralization. For example, in viral studies, detectable neutralization emerges approximately 14.3 days post-symptom onset (range: 3-59 days) .
Isotype contributions: Consider the contributions of different antibody isotypes to neutralization. Early neutralization may occur before detectable IgG responses, suggesting important roles for IgM and IgA in acute settings .
Discrepancies between binding affinity and functional activity of CSLA15 Antibody require careful interpretation:
Epitope accessibility: High binding affinity measured in ELISA may not translate to functional activity if the epitope is poorly accessible in the native protein conformation.
Isotype contributions: Different isotypes may exhibit varying functional capacities despite similar binding. For instance, IgM and IgA may contribute significantly to neutralization even when present at lower concentrations than IgG .
Temporal dynamics: Understand that binding and functional activity may follow different temporal dynamics. Early in immune responses, lower-affinity antibodies may demonstrate functional activity before high-affinity antibodies develop through affinity maturation .
Multiparametric analysis: Analyze correlations between binding (by ELISA) and functional assays (e.g., neutralization tests). Strong correlations support reliability, while discrepancies warrant further investigation of potential mechanisms .
For longitudinal antibody response data analysis:
Mixed-effect models: These account for repeated measures and individual variation, particularly useful when sampling intervals vary between subjects.
Area under the curve (AUC): Calculate AUC to quantify the total antibody response over time, allowing for comparison between groups with different kinetics.
Time-to-event analysis: Determine time to seroconversion or time to reaching specific antibody thresholds using Kaplan-Meier curves and log-rank tests.
Decline rate calculation: For waning antibody responses, calculate the rate of decline after peak responses. Many antibody responses show exponential decay patterns that can be modeled mathematically .
Correlation matrices: Analyze correlations between antibody responses and clinical parameters across multiple timepoints to identify patterns associated with outcomes of interest.
Integrating antibody data with other immunological parameters requires methodological rigor:
Multiparameter correlation analysis: Correlate antibody binding or functional data with cytokine profiles, cellular phenotypes, and clinical outcomes.
Pathway analysis: Examine how CSLA15-related pathways interact with broader immunological networks. For example, in T cell studies, investigate connections between ubiquitin ligase activity (like Cul5) and cytokine signaling pathways that influence T cell differentiation .
Systems serology: Apply systems biology approaches to analyze antibody features (isotype, subclass, glycosylation, Fc receptor binding) alongside functional assays to develop comprehensive immune signatures.
Machine learning approaches: Use dimensionality reduction techniques (PCA, UMAP, t-SNE) to visualize complex immune datasets and identify patterns not apparent in univariate analyses.
Emerging technologies poised to advance CSLA15 Antibody research include:
Single-cell techniques: Technologies like CelliGO that combine high-throughput screening with single-cell sequencing will enable deeper characterization of antibody repertoires and functional properties at unprecedented resolution .
Spatial proteomics: Methods that preserve spatial information while detecting antibody binding will provide insights into the microenvironmental context of target antigen expression.
CRISPR-based validations: Genome editing approaches offer precise validation of antibody specificity by creating knockout controls that eliminate the target epitope.
AI-assisted epitope prediction: Machine learning algorithms increasingly predict antibody-antigen interactions, potentially guiding the development of more specific antibodies with desired functional properties.
Microfluidic technologies: These enable rapid screening of antibody binding and functional properties using minimal sample volumes, accelerating discovery and validation pipelines .
CSLA15 Antibody research may provide valuable insights into T cell fate determination through:
Pathway interrogation: Elucidating the role of ubiquitin ligase complexes (such as Cul5) in regulating cytokine signaling pathways that direct T cell differentiation into distinct functional subsets .
Tolerance mechanisms: Investigating how specific molecular pathways promote tolerogenic T cell fate choices, potentially revealing targets for therapeutic intervention in allergic and autoimmune diseases .
Cytokine signal modulation: Exploring how post-translational modifications, particularly ubiquitination, regulate the intensity and duration of cytokine signaling to influence T cell differentiation toward inflammatory (Th2, Th9) versus regulatory (Treg) phenotypes .
Transcriptional network analysis: Mapping how signaling events regulated by ubiquitin ligases connect to transcriptional networks that establish and maintain T cell identity and function .