TARC Mouse

Thymus and Activation Regulated Chemokine (CCL17) Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Characterization

Mouse TARC is encoded by a 93-amino-acid precursor protein, processed into a mature 70-amino-acid peptide (7.9 kDa) after cleavage of its 23-residue signal peptide . Key features include:

PropertyDetails
Amino Acid Identity64% with human TARC; 83% with rat TARC
Production MethodRecombinant expression in E. coli (non-glycosylated, 0.2 µm filtered)
StabilityStable at -18°C (lyophilized); 4°C for short-term storage

Biological Roles and Mechanisms

TARC binds CCR4 and CCR8 receptors, directing immune cell migration and modulating inflammation:

Key Functions

  • Chemotaxis: Attracts CCR4+ T cells, dendritic cells (DCs), and Th2 lymphocytes .

  • Immune Regulation:

    • Upregulated by IL-4 and TNF-α in Langerhans cells .

    • Inhibited by IFN-γ in macrophages and monocytes .

  • Disease Associations:

    • Asthma: Elevated in bronchial epithelium and peribronchial lesions post-antigen challenge .

    • Autoimmunity: Higher plasma levels in NZB/W F1 lupus-prone mice .

Regulatory Factors

InducersInhibitorsCell Types
IL-4, TNF-α, IL-13IFN-γ, IL-10Dendritic cells
GM-CSF, RANK engagementGlucocorticoids (no effect)Macrophages

Experimental Models

  • Asthma:

    • OVA-challenged mice showed 3–24h post-exposure TARC mRNA peaks in lung tissue, correlating with eosinophil infiltration .

    • Anti-TARC monoclonal antibodies (e.g., 5H5) neutralized chemotaxis (ND₅₀: 0.2–0.6 µg/mL) .

  • Biomarker Potential:

    • Serum TARC >269.9 pg/mL distinguished IgG4-related disease from Sjögren’s syndrome (AUC: 0.97) .

Therapeutic Targeting

  • Neutralizing TARC reduced airway hyperresponsiveness (AHR) and eosinophilia in murine asthma models .

Detection and Quantification

The Quantikine® Mouse TARC Immunoassay (R&D Systems MCC170) is widely used:

ParameterIntra-Assay CV%Inter-Assay CV%
Low (27–29 pg/mL)4.87.6
Medium (89–98 pg/mL)2.67.0
High (278–310 pg/mL)2.85.6

Validated for serum, plasma, and cell culture supernates .

Product Specs

Introduction
TARC cDNA encodes a 94 amino acid precursor protein. After cleavage of a 23 amino acid signal peptide, the mature secreted protein of 71 amino acids is generated. TARC shares approximately 24-29% amino acid sequence identity with CC chemokine family members such as RANTES, MIP-1a, MIP-1b, MCP-1, MCP-2, MCP-3 and I-309. Expression of TARC is observed in the thymus, with lower levels found in the lung, colon, and small intestine. Transient expression can also be detected in stimulated peripheral blood mononuclear cells. Recombinant TARC exhibits chemotactic activity for T cell lines, but not for monocytes or neutrophils. As a specific functional ligand for CCR4, a receptor selectively expressed on T cells, CCL-17 plays a crucial role. CCL17 is among a group of Cys-Cys (CC) cytokine genes clustered on chromosome 16q. It displays chemotactic activity for T lymphocytes, but not monocytes or granulocytes. CCL17 binds to chemokine receptors CCR4 and CCR8. This chemokine plays a vital role in T cell development in the thymus, as well as in the trafficking and activation of mature T cells.
Description
Recombinant Mouse TARC, produced in E. coli, is a non-glycosylated polypeptide chain comprising 70 amino acids with a molecular weight of 7.9 kDa. The purification process involves proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
The protein was lyophilized from a 0.2 µm filtered solution in 1xPBS, pH 7.4.
Solubility
To reconstitute the lyophilized TARC, it is recommended to dissolve it in sterile 18 MΩ-cm H₂O at a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized TARC remains stable at room temperature for up to 3 weeks. However, it is recommended to store it desiccated below -18°C. After reconstitution, store TARC at 4°C for 2-7 days. For long-term storage, it is advisable to store it below -18°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 97.0% as determined by: (a) RP-HPLC analysis. (b) SDS-PAGE analysis.
Biological Activity
The ED50, determined by the chemoattractant activity on BaF3 mouse pro-B cells transfected with human CCR4, is typically between 2-10 ng/ml.
Synonyms
C-C motif chemokine 17, Small-inducible cytokine A17, Thymus and activation-regulated chemokine, CC chemokine TARC, ABCD-2, CCL17, CCL-17, SCYA17, TARC, A-152E5.3, MGC138271, MGC138273.
Source
Escherichia Coli.
Amino Acid Sequence
ARATNVGREC CLDYFKGAIP IRKLVSWYKT SVECSRDAIV FLTVQGKLIC ADPKDKHVKK AIRLVKNPRP.

Q&A

What is TARC/CCL17 and how is it expressed in mouse tissues?

TARC/CCL17 is a CC chemokine that was identified using a signal sequence trap method. In mice, it encodes a highly basic 94 amino acid precursor protein with a 23 amino acid signal peptide that is cleaved to generate a 71 amino acid mature secreted protein . TARC/CCL17 exhibits tissue-specific expression patterns, being constitutively expressed in the thymus and at lower levels in the lung, colon, and small intestine . It can also be transiently expressed in stimulated peripheral blood mononuclear cells.

Immunohistochemical studies have demonstrated that TARC is consistently present in the thymus regardless of experimental conditions, while its expression in skin tissues can be significantly induced under specific conditions such as drug exposure . For instance, in abacavir-treated mice, TARC was observed in ear skin but was barely detectable in vehicle-treated controls .

How does mouse TARC/CCL17 compare structurally to other chemokines?

Mouse TARC/CCL17 shares approximately 24-29% amino acid sequence identity with other CC chemokine family members, including RANTES, MIP-1α, MIP-1β, MCP-1, MCP-2, MCP-3, and I-309 . This relatively low sequence homology contributes to TARC's distinct functional properties and receptor specificity. Despite these differences, TARC/CCL17 maintains the characteristic structural features of CC chemokines that enable it to interact with specific receptors, primarily CCR4 and CCR8 .

The structural distinctiveness of TARC/CCL17 is important for researchers developing specific antibodies for experimental applications. For example, the anti-mouse TARC monoclonal antibody 5H5 demonstrates high specificity, binding only to recombinant mouse TARC protein but not to other tested mouse CC chemokines, including MDC, liver and activation-regulated chemokine/MIP-3α, and several others .

What are the primary cellular sources of TARC/CCL17 in mice?

In mouse models, TARC/CCL17 is produced by multiple cell types within the immune system and barrier tissues. The primary cellular sources include:

  • Thymic dendritic cells

  • Lymph node dendritic cells

  • Monocytes

  • CD4+ T cells

  • Keratinocytes

  • Fibroblasts

  • Bronchial epithelial cells

The production can be constitutive (as in thymic tissues) or induced, depending on the tissue and physiological context. For example, Reed-Sternberg cells have also been identified as producers of TARC/CCL17 . This diverse cellular expression pattern contributes to TARC's multifaceted roles in immune regulation and inflammatory responses across different tissues and disease states.

What role does TARC/CCL17 play in mouse inflammatory responses?

TARC/CCL17 functions primarily by interacting with CCR4 and CCR8 receptors to induce the chemoattraction of activated Th2 cells, basophils, and NK cells to sites of inflammation . Through these interactions, TARC/CCL17 promotes:

  • Th2 cell recruitment to inflammatory sites

  • Induction of platelet aggregation and degranulation

  • Modulation of allergic and inflammatory responses

In mouse models of allergic inflammation, neutralization of TARC/CCL17 attenuates the development of allergic airway inflammation and hyperresponsiveness , demonstrating its integral role in these processes. The regulation of TARC/CCL17 appears to involve CD4+ T cells, as studies have shown that CD4+ T cell depletion affects TARC/CCL17 expression patterns in experimental models .

What methods are most effective for measuring TARC/CCL17 in mouse samples?

For quantitative assessment of TARC/CCL17 in mouse samples, enzyme-linked immunosorbent assay (ELISA) represents the gold standard approach. Commercial kits such as the Mouse CCL17/TARC Quantikine ELISA Kit use E. coli-expressed recombinant mouse TARC and specific antibodies for reliable detection . When implementing ELISA-based measurements, researchers should consider the following performance characteristics:

ParameterCell Culture SupernatesSerumEDTA Plasma
Intra-Assay CV%2.6-4.82.6-4.82.6-4.8
Inter-Assay CV%5.6-7.65.6-7.65.6-7.6
Recovery %94 (85-113)94 (82-103)94 (83-109)

The detection threshold for these assays is typically in the picogram/ml range, making them suitable for detecting physiologically relevant concentrations . For values below the detectable range, researchers typically use the lower limit of quantification for statistical analyses .

For tissue-specific TARC/CCL17 expression patterns, immunohistochemistry (IHC) provides valuable spatial information. This approach has successfully revealed differential expression in tissues such as thymus, skin, liver, and spleen under various experimental conditions .

What are the validated protocols for TARC/CCL17 neutralization in mice?

TARC/CCL17 neutralization in mouse models can be achieved using specific monoclonal antibodies. A validated protocol based on published research includes:

  • Antibody selection: Anti-mouse TARC monoclonal antibody (such as clone 110904 from R&D Systems) or the highly specific neutralizing mAb 5H5, which demonstrates exclusive binding to recombinant mouse TARC protein without cross-reactivity to other chemokines .

  • Administration schedule: Intraperitoneal (i.p.) injection of anti-TARC mAb at 20 μg/body on days -1, 1, 3, and 5 of the experimental timeline .

  • Control implementation: Concurrent administration of isotype control immunoglobulin G (IgG) to control animals .

  • Validation of neutralization efficacy: Functional assays such as calcium mobilization and chemotaxis assays in mouse cells expressing CCR4 can confirm successful neutralization .

Importantly, anti-TARC mAb treatment has been demonstrated to be functionally equivalent to TARC-deficiency in mice , providing researchers with a flexible alternative to genetic knockout approaches for studying TARC/CCL17 functions.

How do researchers establish the specificity of anti-TARC antibodies in mouse studies?

Establishing antibody specificity is critical for reliable TARC/CCL17 research. A comprehensive approach includes:

  • Direct ELISA binding assessment: Testing antibody binding to recombinant mouse TARC protein versus other mouse CC chemokines. For example, the mAb 5H5 has been verified to bind only to TARC and not to MDC, liver and activation-regulated chemokine/MIP-3α, secondary lymphoid chemokine/6Ckine, EBI1-ligand chemokine/MIP-3β, stromal-derived factor-1, RANTES, lymphotactin, MIP-1α, MCP-1, or IL-11 receptor α locus chemokine .

  • Functional inhibition assays: Verifying that the antibody inhibits TARC-specific cellular responses without affecting responses to related chemokines. This can be assessed through:

    • Calcium mobilization assays in CCR4-expressing cells

    • Chemotaxis assays using stably transfected cell lines

  • Cross-reactivity testing: Systematic evaluation against structurally or functionally related molecules, particularly MDC (CCL22), which also binds to CCR4.

  • In vivo validation: Confirming that antibody administration produces effects consistent with TARC deficiency in established mouse models.

For instance, the 5H5 antibody completely inhibited mouse TARC-induced calcium mobilization and chemotaxis in CCR4-expressing cells but had no effect on mouse MDC-induced responses, confirming its high specificity .

How should researchers interpret serum TARC/CCL17 levels in different mouse models?

Interpreting serum TARC/CCL17 levels requires careful consideration of several factors:

  • Baseline variation: Normal serum TARC levels differ between mouse strains and experimental conditions. Establishing appropriate baselines for each model is essential.

  • Disease correlation: In specific models, such as abacavir hypersensitivity syndrome (AHS), serum TARC levels correlate with disease severity. For example, transgenic mice receiving abacavir showed elevated serum TARC levels compared to controls, with further increases when combined with CD4+ T cell depletion .

  • Statistical handling: For values below the detectable range, consistent approaches such as using the lower limit of quantification should be implemented .

  • Contextual analysis: Changes in serum TARC should be interpreted alongside tissue-specific expression patterns, which may reveal important discrepancies. For instance, while serum TARC levels increase in certain conditions, tissue expression might show organ-specific patterns, as observed in studies where TARC was detected in skin but not in liver or spleen of treated mice .

  • Treatment response: In intervention studies, changes in serum TARC can serve as biomarkers for treatment efficacy, particularly in allergic and inflammatory models .

How does TARC/CCL17 contribute to allergic airway inflammation in mouse models?

TARC/CCL17 plays multiple critical roles in allergic airway inflammation through several mechanisms:

  • T cell recruitment: By interacting with CCR4 and CCR8, TARC/CCL17 mediates the recruitment of activated Th2 cells to the airways, a key event in allergic inflammation .

  • Inflammatory cascade regulation: TARC/CCL17 facilitates the production of Th2 cytokines including IL-4 and IL-13, which are central to allergic responses. Neutralizing TARC can reduce these cytokines in bronchoalveolar lavage fluid .

  • Airway hyperresponsiveness modulation: Intervention studies using anti-TARC antibodies have demonstrated that TARC neutralization attenuates the development of both allergic airway inflammation and hyperresponsiveness in mice .

  • Cellular recruitment beyond T cells: TARC/CCL17 contributes to the recruitment of other inflammatory cells, including basophils and NK cells, amplifying the allergic response .

These findings position TARC/CCL17 as a potential therapeutic target for allergic airway diseases, with neutralization strategies showing promising results in preclinical models.

How does TARC/CCL17 expression correlate with skin inflammation in mouse models?

The relationship between TARC/CCL17 expression and skin inflammation in mouse models is characterized by:

  • Inducible expression pattern: While TARC/CCL17 is barely detectable in normal mouse skin, it becomes significantly upregulated under inflammatory conditions. Immunohistochemical studies have shown that in abacavir-treated mice, TARC was clearly observed in ear skin but remained almost undetectable in vehicle-treated controls .

  • Severity correlation: Research indicates that TARC expression is induced in mouse models of skin toxicity, with the severity of cutaneous reactions positively correlating with serum TARC levels .

  • T cell dynamics: TARC/CCL17 expression in skin appears to be associated with T cell-mediated inflammation, particularly in drug hypersensitivity models. Studies have investigated whether TARC expression correlates with CD8+ T cell infiltration in these contexts .

  • Biomarker potential: The strong correlation between skin inflammation and TARC/CCL17 levels suggests its utility as a biomarker for monitoring skin inflammatory conditions in experimental models and potentially in translational research.

This relationship between TARC/CCL17 and skin inflammation provides insights into the pathogenesis of cutaneous inflammatory disorders and suggests potential therapeutic approaches targeting this chemokine.

What methodological approaches are most effective for studying TARC/CCL17 in tissue-specific contexts?

For comprehensive analysis of TARC/CCL17 in tissue-specific contexts, researchers should employ a multi-faceted approach:

  • Tissue expression mapping:

    • Immunohistochemistry (IHC) for spatial localization within tissues, as demonstrated in studies examining TARC expression in thymus, liver, spleen, and ear skin

    • In situ hybridization for mRNA detection with cellular resolution

    • Laser capture microdissection combined with qPCR for precise anatomical analysis

  • Quantitative assessment:

    • ELISA of tissue homogenates for protein quantification

    • qRT-PCR for mRNA expression analysis

    • Western blotting for protein expression with size confirmation

  • Functional studies:

    • Tissue-specific conditional knockout models

    • Local administration of neutralizing antibodies

    • Ex vivo tissue explant cultures with TARC/CCL17 modulation

  • Cellular source identification:

    • Flow cytometry of tissue-derived cell suspensions

    • Single-cell RNA sequencing for unbiased profiling

    • Cell sorting followed by TARC/CCL17 expression analysis

  • Receptor interaction studies:

    • Receptor antagonist administration to specific tissues

    • Fluorescently labeled TARC/CCL17 for binding visualization

    • Calcium flux assays in tissue-derived cells

These methodological approaches, when applied systematically, can reveal tissue-specific nuances of TARC/CCL17 biology that might be missed by focusing solely on systemic measurements or single analytical techniques.

How do genetic and pharmacological approaches to TARC/CCL17 inhibition compare in mouse models?

Comparing genetic and pharmacological approaches to TARC/CCL17 inhibition reveals important distinctions and complementary insights:

Genetic Approaches:

  • Complete elimination of TARC/CCL17 expression when using germline knockouts

  • Temporal control possible with inducible knockout systems

  • Cell type-specific deletion achievable with conditional knockouts

  • Provides insights into developmental and compensatory mechanisms

  • May reveal unexpected phenotypes due to complete absence throughout development

Pharmacological Approaches:

  • Dosage-dependent inhibition allowing titration of effect

  • Temporal flexibility with administration at any experimental timepoint

  • Potentially fewer compensatory mechanisms compared to genetic deletion

  • More directly translatable to therapeutic applications

  • Validated protocols include anti-TARC mAb administration (20 μg/body, i.p.) on days -1, 1, 3, and 5

Importantly, studies have demonstrated that anti-TARC monoclonal antibody treatment appears functionally equivalent to TARC-deficiency in mice , suggesting that for many research questions, the pharmacological approach provides a reliable alternative to genetic models. This equivalence facilitates experimental design flexibility while maintaining physiological relevance.

The choice between these approaches should be guided by the specific research question, with combined approaches often providing the most comprehensive understanding of TARC/CCL17 biology in disease models.

How can researchers address variability in TARC/CCL17 measurements across mouse samples?

Variability in TARC/CCL17 measurements represents a common challenge that can be methodically addressed through several strategies:

  • Standardized sample handling:

    • Implement consistent timing for sample collection

    • Standardize processing protocols, including clotting time for serum

    • Minimize freeze-thaw cycles

    • Use consistent anticoagulants for plasma samples

  • Technical optimization:

    • Perform technical replicates (at minimum duplicates) based on known assay variability (Intra-Assay CV of 2.6-4.8% and Inter-Assay CV of 5.6-7.6%)

    • Validate recovery efficiency in each matrix (94% recovery reported for cell culture supernatants, serum, and EDTA plasma)

    • Ensure consistent sample dilutions within the linear range of the assay

  • Experimental design considerations:

    • Calculate appropriate sample sizes based on expected variability

    • Include age-matched and sex-matched controls

    • Consider circadian variations in chemokine expression

    • Account for housing conditions and environmental factors

  • Statistical approaches:

    • Apply appropriate statistical methods based on data distribution

    • Handle values below detection limits consistently (e.g., using the lower limit of quantification)

    • Consider non-parametric methods when distributions are non-normal

    • Include potential confounding variables in statistical models

  • Validation strategies:

    • Confirm key findings using alternative detection methods

    • Correlate serum measurements with tissue expression where relevant

    • Consider functional assays to validate biological significance

What controls are essential when studying TARC/CCL17 neutralization in mouse models?

Robust experimental design for TARC/CCL17 neutralization studies requires several critical controls:

  • Antibody controls:

    • Isotype control antibodies matching the neutralizing antibody class (e.g., IgG2A for anti-TARC mAb)

    • Dose-matched control antibody administration

    • Verification of antibody specificity through binding assays

    • Functional validation through calcium mobilization and chemotaxis inhibition

  • Treatment controls:

    • Vehicle controls (e.g., phosphate-buffered saline) for antibody administration

    • Matched administration schedule and route

    • Handling controls to account for stress effects

  • Experimental validation:

    • Positive controls (known TARC/CCL17-dependent models)

    • Dose-response assessment for neutralizing antibodies

    • Temporal controls examining timing of neutralization

  • Analytical controls:

    • Standard curves for quantitative assays

    • Matrix-matched calibrators

    • Reference samples across experimental batches

  • Biological controls:

    • Wild-type littermates for genetic models

    • Age-matched and sex-matched subjects

    • Health status monitoring

Particularly important is confirming antibody specificity, as demonstrated with the 5H5 anti-TARC mAb, which was validated through direct ELISA to bind only to recombinant mouse TARC protein and not to other tested mouse CC chemokines . Additionally, functional validation through inhibition of TARC-induced but not MDC-induced calcium mobilization and chemotaxis provides crucial evidence of specificity .

How should researchers address contradictory findings in TARC/CCL17 expression across different mouse models?

When encountering contradictory findings regarding TARC/CCL17 expression, researchers should implement a systematic approach:

  • Methodological reconciliation:

    • Compare detection methods (ELISA vs. IHC vs. PCR)

    • Evaluate antibody specificities and recognition epitopes

    • Consider detection thresholds and dynamic ranges

    • Examine sample preparation differences

  • Model-specific considerations:

    • Analyze genetic background differences between mouse strains

    • Evaluate age-dependent expression patterns

    • Consider disease model variations (e.g., acute vs. chronic)

    • Examine intervention protocols and timing

  • Tissue-specific analysis:

    • Recognize differential expression patterns (e.g., constitutive in thymus vs. inducible in skin)

    • Compare local vs. systemic expression

    • Consider micro-environmental factors influencing expression

  • Biological validation:

    • Implement multiple complementary detection methods

    • Correlate expression with functional outcomes

    • Perform time-course studies to capture dynamic changes

  • Experimental design refinement:

    • Increase biological replicates

    • Include positive and negative controls

    • Standardize experimental conditions

For example, reconciling observations that TARC exists in thymus regardless of experimental conditions while being inducible in skin requires recognizing tissue-specific regulation rather than viewing these as contradictory findings. Similarly, understanding that serum TARC levels may not directly reflect tissue expression patterns helps explain apparent discrepancies between systemic and local measurements.

What factors should be considered when analyzing TARC/CCL17 data in correlation with disease severity?

When analyzing correlations between TARC/CCL17 expression and disease severity, several critical factors require consideration:

  • Temporal relationship:

    • Determine whether TARC/CCL17 changes precede, coincide with, or follow disease manifestations

    • Implement longitudinal sampling when possible

    • Consider kinetic differences between protein expression and clinical manifestations

  • Dose-response assessment:

    • Evaluate whether correlations are linear or threshold-dependent

    • Determine if correlations persist across the full spectrum of disease severity

    • Consider ceiling or floor effects in detection methods

  • Confounding variables:

    • Account for treatment effects on both TARC/CCL17 and disease markers

    • Control for demographic variables (age, sex, genetic background)

    • Consider comorbid conditions or secondary manifestations

  • Multivariate analysis:

    • Include other relevant biomarkers in correlation models

    • Apply appropriate statistical methods for multiple correlations

    • Consider principle component analysis for complex datasets

  • Causal relationship investigation:

    • Test whether TARC/CCL17 modulation affects disease course

    • Evaluate whether disease modification affects TARC/CCL17 levels

    • Implement conditional knockout or neutralization studies

For example, studies have identified positive correlations between serum TARC levels and skin toxicity severity in mouse models . To fully interpret such correlations, researchers should determine whether interventions targeting TARC/CCL17 ameliorate skin manifestations, as this would strengthen the case for a causal rather than merely associative relationship.

What emerging technologies might advance TARC/CCL17 research in mouse models?

Several cutting-edge technologies hold promise for advancing TARC/CCL17 research:

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify precise cellular sources of TARC/CCL17

    • Single-cell proteomics for protein-level characterization

    • Spatial transcriptomics to map expression with tissue context preservation

  • Advanced imaging approaches:

    • Intravital microscopy for real-time visualization of TARC/CCL17-mediated cell recruitment

    • Multiplexed immunofluorescence for simultaneous detection of multiple markers

    • Light-sheet microscopy for whole-organ imaging with cellular resolution

  • Genetic engineering advancements:

    • CRISPR/Cas9-based approaches for precise genetic modifications

    • Inducible and reversible gene regulation systems

    • Cell type-specific promoters for targeted expression studies

  • Systems biology tools:

    • Multi-omics integration platforms

    • Network analysis of TARC/CCL17 in immune signaling pathways

    • Machine learning approaches for pattern recognition in complex datasets

  • Nanobody and aptamer technologies:

    • Development of smaller binding molecules for improved tissue penetration

    • Bispecific constructs targeting TARC/CCL17 and its receptors simultaneously

    • Targeted degradation approaches using proteolysis-targeting chimeras (PROTACs)

These technologies would enable researchers to address existing knowledge gaps, such as the precise cellular dynamics of TARC/CCL17 production in different disease states and the temporal relationship between TARC/CCL17 expression and disease progression.

What are the most promising therapeutic applications of TARC/CCL17 research in mouse models?

TARC/CCL17 research in mouse models reveals several promising therapeutic applications:

  • Allergic airway diseases:

    • Intervention studies have demonstrated that TARC/CCL17 neutralization attenuates allergic airway inflammation and hyperresponsiveness

    • Potential applications in asthma and allergic rhinitis

    • Combination approaches with existing therapies

  • Inflammatory skin conditions:

    • Strong correlations between TARC/CCL17 expression and skin inflammation severity

    • Potential applications in atopic dermatitis, drug hypersensitivity reactions, and psoriasis

    • Topical and systemic delivery approaches

  • Drug hypersensitivity syndromes:

    • TARC/CCL17 serves as a biomarker in models of abacavir hypersensitivity

    • Potential for early detection and intervention in adverse drug reactions

    • Personalized medicine approaches based on TARC/CCL17 profiles

  • Th2-driven inflammatory disorders:

    • TARC/CCL17 promotes recruitment of Th2 cells to inflammatory sites

    • Applications in eosinophilic disorders and allergic inflammation

    • Targeted therapies disrupting the TARC/CCL17-CCR4/CCR8 axis

  • Biomarker applications:

    • Use of serum TARC/CCL17 levels for disease monitoring

    • Stratification of patients for clinical trials

    • Prediction of treatment response

The translation of these findings to human applications will require careful validation of cross-species conservation of mechanisms and thorough safety assessments, particularly given TARC/CCL17's physiological roles in normal immune function.

What methodological gaps need to be addressed in TARC/CCL17 mouse research?

Despite significant advances, several methodological gaps remain in TARC/CCL17 research:

  • Standardization challenges:

    • Lack of uniform protocols for sample collection and processing

    • Variability in detection methods and reporting formats

    • Need for reference standards and quality control materials

  • Temporal dynamics:

    • Limited understanding of circadian and developmental regulation

    • Insufficient longitudinal studies with sequential sampling

    • Need for real-time monitoring technologies

  • Tissue accessibility:

    • Challenges in detecting TARC/CCL17 in certain tissues

    • Limited methods for non-terminal sampling in longitudinal studies

    • Need for improved in vivo imaging approaches

  • Functional assessment:

    • Over-reliance on concentration measurements rather than activity assays

    • Limited tools for distinguishing active versus inactive forms

    • Need for receptor-specific functional readouts

  • Translational barriers:

    • Incomplete understanding of mouse-to-human differences

    • Limited validation in humanized mouse models

    • Need for better predictive markers of human responses

Addressing these gaps would enhance the reliability and translational value of TARC/CCL17 research. Particularly valuable would be the development of standardized protocols for tissue-specific TARC/CCL17 detection and functional assessment, along with improved methods for longitudinal monitoring in individual animals.

How might TARC/CCL17 interact with other chemokine systems in complex disease models?

Understanding TARC/CCL17 interactions with other chemokine systems represents a frontier in chemokine research:

  • Receptor competition and cooperation:

    • TARC/CCL17 shares receptors (CCR4, CCR8) with other chemokines, creating potential competition or synergy

    • The balance between TARC/CCL17 and MDC (CCL22) at the CCR4 receptor may determine biological outcomes

    • Heterodimer formation between chemokines may create novel signaling properties

  • Temporal coordination:

    • Sequential chemokine expression may create a choreographed immune response

    • TARC/CCL17 may initiate responses that are amplified or resolved by other chemokines

    • The kinetics of receptor responsiveness may evolve during disease progression

  • Spatial organization:

    • Chemokine gradients may work cooperatively to guide cell migration

    • Tissue-specific expression patterns may create microanatomical domains

    • Local production versus systemic circulation creates overlapping gradients

  • Functional redundancy and specialization:

    • Multiple chemokines may provide backup systems for critical immune functions

    • Specific combinations may be required for optimal responses

    • Disease states may dysregulate the normal balance between chemokine systems

  • Therapeutic implications:

    • Targeting multiple chemokines simultaneously may provide synergistic benefits

    • Compensatory upregulation may limit efficacy of single-target approaches

    • Chemokine network analysis may identify optimal intervention points

Advanced approaches combining multi-parameter measurement with systems biology analysis will be essential for unraveling these complex interactions and identifying the most promising therapeutic strategies targeting TARC/CCL17 in the context of the broader chemokine network.

Product Science Overview

Introduction

Thymus and Activation Regulated Chemokine (CCL17), also known as TARC, is a member of the CC chemokine family. It is a small cytokine that plays a crucial role in the immune system by mediating the migration of immune cells to sites of inflammation or injury. CCL17 is particularly significant in the context of allergic reactions and autoimmune diseases.

Gene and Protein Structure

The CCL17 gene is located on chromosome 8 in mice . The protein encoded by this gene is a chemokine that displays chemotactic activity specifically for T lymphocytes, but not for monocytes or granulocytes . The protein binds to chemokine receptors CCR4 and CCR8, which are expressed on various immune cells .

Expression and Regulation

CCL17 is constitutively expressed in the thymus and can be induced in several cell types upon activation . It is produced by antigen-presenting cells such as dendritic cells, macrophages, and monocytes . The expression of CCL17 is regulated by various cytokines and inflammatory signals, making it a key player in the immune response.

Biological Functions

CCL17 plays a pivotal role in T cell development in the thymus as well as in the trafficking and activation of mature T cells . It is involved in the recruitment of Th2 cells and CLA+ CD4+ T cells, which are essential for the immune response in allergic diseases . CCL17 also mediates chemotaxis, the directed movement of cells towards higher concentrations of the chemokine, thereby facilitating the migration of immune cells to sites of inflammation .

Role in Diseases

CCL17 has been implicated in various diseases, particularly those involving the immune system. It plays a complex role in cancer, where it can attract T-regulatory cells, allowing some cancers to evade an immune response . Conversely, in other cancers such as melanoma, an increase in CCL17 is linked to improved outcomes . Additionally, CCL17 is associated with autoimmune and allergic diseases, including atopic dermatitis, allergic asthma, allergic rhinitis, and allergic contact dermatitis .

Recombinant CCL17

Recombinant CCL17 (Mouse) is a laboratory-produced version of the natural chemokine. It is used in research to study the functions and mechanisms of CCL17 in various biological processes and diseases. Recombinant proteins are typically produced using bacterial or mammalian expression systems, allowing for the generation of large quantities of the protein for experimental purposes.

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.