CCL17 binds to receptors CCR4 and CCR8, driving immune responses through:
T-cell chemotaxis: Directs migration of Th2 cells, regulatory T cells (Tregs), and skin-homing T cells .
Microglial modulation: Enhances phagocytosis in microglia via CCR4/ERK/Nrf2 pathways, promoting hematoma resolution after intracerebral hemorrhage (ICH) .
Inflammatory amplification: Upregulates CCL2 in macrophages, exacerbating cell accumulation in chronic obstructive pulmonary disease (COPD) models .
In rats, endogenous CCL17 peaks 72 hours post-ICH, correlating with CD163+ microglial activation and improved neurological outcomes .
ICH recovery: Intranasal recombinant CCL17 (rCCL17) in mice reduced hematoma volume by 40% and improved motor function via CCR4/ERK/Nrf2 signaling .
Subarachnoid hemorrhage (SAH): rCCL17 attenuated brain edema and promoted M2-like microglial polarization through CCR4/mTORC2 pathways .
COPD models: CCL17 increased macrophage recruitment in bronchoalveolar lavage fluid (BALF) by 2.5-fold, synergizing with cigarette smoke to worsen emphysema .
Autoimmunity: Ccl17 knockout mice showed reduced inflammation in allergic and autoimmune disorders .
Neuroprotection: Targeting CCR4 with rCCL17 may mitigate early brain injury post-hemorrhage .
Cancer immunotherapy: Dual role observed—CCL17 recruits Tregs to suppress anti-tumor immunity but enhances dendritic cell efficacy in melanoma .
Autoimmune disease: Inhibiting CCL17/CCR4 axis reduces inflammation in rheumatoid arthritis and asthma models .
Recombinant rat CCL17 protein is produced through a robust process. The gene fragment encoding the 24-93 amino acid residues of rat CCL17 is cloned into a vector and inserted into E. coli. This is followed by upstream bioprocessing and downstream purification steps. The final product exhibits a purity exceeding 97% as determined by SDS-PAGE and contains endotoxin levels below 1.0 EU/µg as measured by the LAL method. This recombinant rat CCL17 protein has been validated for its biological activity, demonstrating a concentration-dependent chemotaxis response in human T-lymphocytes within the range of 1.0-10 ng/ml.
Rat CCL17 plays a critical role in the recruitment and activation of Th2 cells, which are key mediators of allergic responses and various inflammatory conditions. This chemokine is constitutively expressed in the thymus and its expression can be induced in various cell types, including peripheral blood mononuclear cells, macrophages, and bronchial epithelial cells [1]. In rat models, CCL17 has been implicated in several pathophysiological conditions, including asthma, pulmonary fibrosis, and responses to environmental pollutants such as cigarette smoke [1][2][3].
The interaction of CCL17 with its receptor, CCR4, is crucial in mediating Th2 cell responses. Studies have shown that CCL17 expression is upregulated in the lungs of rats exposed to various inflammatory stimuli, coinciding with an increase in CCR4-positive lymphocytes, suggesting a role in Th2-dominant immune responses [2][3]. Furthermore, neutralization of CCL17 has been shown to reduce airway hyperresponsiveness in animal models, highlighting its importance in asthma pathogenesis [4][5].
Research has also indicated that CCL17 is involved in the pathophysiology of idiopathic pulmonary fibrosis (IPF). Elevated levels of CCL17 and CCR4 have been observed in models of bleomycin-induced pulmonary fibrosis, emphasizing its role in the recruitment of inflammatory cells to the lung tissue [3].
References:
[1] S. Suzuki, K. Asai, M. Gi, K. Kojima, A. Kakehashi, Y. Oishiet al., Response biomarkers of inhalation exposure to cigarette smoke in the mouse lung, Journal of Toxicologic Pathology, vol. 35, no. 3, p. 247-254, 2022. https://doi.org/10.1293/tox.2021-0077
[2] T. Inoue, S. Fujishima, E. Ikeda, O. Yoshie, N. Tsukamoto, S. Aisoet al., Ccl22 and ccl17 in rat radiation pneumonitis and in human idiopathic pulmonary fibrosis, European Respiratory Journal, vol. 24, no. 1, p. 49-56, 2004. https://doi.org/10.1183/09031936.04.00110203
[3] Y. Yogo, S. Fujishima, T. Inoue, F. Saito, T. Shiomi, K. Yamaguchiet al., Macrophage derived chemokine (ccl22), thymus and activation-regulated chemokine (ccl17), and ccr4 in idiopathic pulmonary fibrosis, Respiratory Research, vol. 10, no. 1, 2009. https://doi.org/10.1186/1465-9921-10-80
[4] S. Santulli-Marotto, K. Boakye, E. Lacy, S. Wu, J. Luongo, K. Kavalkovichet al., Engagement of two distinct binding domains on ccl17 is required for signaling through ccr4 and establishment of localized inflammatory conditions in the lung, Plos One, vol. 8, no. 12, p. e81465, 2013. https://doi.org/10.1371/journal.pone.0081465
[5] S. Santulli-Marotto, J. Fisher, T. Petley, K. Boakye, T. Panavas, J. Luongoet al., Surrogate antibodies that specifically bind and neutralize ccl17 but not ccl22, Monoclonal Antibodies in Immunodiagnosis and Immunotherapy, vol. 32, no. 3, p. 162-171, 2013. https://doi.org/10.1089/mab.2012.0112
Rat CCL17 adopts the canonical chemokine fold consisting of three β-strands and a C-terminal α-helix. The N-terminal region preceding the first cysteine residue is critical for receptor activation, while the N-loop region (following the CC motif) is essential for receptor binding specificity. The presence of two disulfide bonds (C11-C35 and C12-C51) stabilizes the tertiary structure, which is crucial for maintaining the spatial orientation of the receptor-binding domains. This precise structural arrangement enables specific interaction with CCR4 receptors on target cells, facilitating chemotaxis of CCR4-expressing cells at concentrations of 1.0-10 ng/ml .
For optimal bioactivity maintenance:
Reconstitution protocol:
Centrifuge the vial briefly before opening to collect contents at the bottom
Reconstitute the lyophilized protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL
Add glycerol to a final concentration of 5-50% (optimally 50%)
Aliquot to minimize freeze-thaw cycles
Storage conditions:
Long-term storage: -20°C/-80°C in aliquots
Working aliquots: 4°C for up to one week
Avoid repeated freeze-thaw cycles which reduce activity
The reconstituted protein maintains stability for up to 6 months at -20°C/-80°C when properly aliquoted with glycerol as a cryoprotectant . The lyophilized form is shipped with buffer components from a 0.2 μm filtered PBS, pH 7.4, which provides optimal stability during reconstitution.
Bioactivity of recombinant Rat CCL17 can be assessed through multiple complementary approaches:
Primary method - Chemotaxis bioassay:
Isolate human or rat T-lymphocytes using density gradient centrifugation
Prepare serial dilutions of CCL17 (1.0-10 ng/ml) in chemotaxis buffer
Place 25-50 μl of cell suspension (1-2×10⁶ cells/ml) in the upper chamber of a transwell system
Add CCL17 dilutions to the lower chamber
Incubate at 37°C with 5% CO₂ for 2-3 hours
Count migrated cells using flow cytometry or hemocytometer
Calculate chemotaxis index: (migrated cells with chemokine)/(migrated cells without chemokine)
Active CCL17 typically induces significant chemotaxis in the 1.0-10 ng/ml concentration range .
Secondary validation methods:
Calcium flux assay in CCR4-expressing cells
Phosphorylation of downstream signaling molecules (e.g., ERK1/2, AKT)
Receptor internalization assay using CCR4-expressing cells
These complementary approaches provide comprehensive validation of functional activity beyond simple binding assays.
To distinguish CCL17-specific effects from other chemokine activities:
Genetic approaches:
Use CCL17E/E knockout models, where the CCL17 gene is replaced with a fluorescent reporter (EGFP)
Employ CCR4-deficient models to eliminate the primary receptor response
Create conditional cell-specific CCL17 knockout models using Cre-loxP systems
Pharmacological approaches:
Use CCR4-specific antagonists such as AZD2098 to block CCL17 signaling
Apply neutralizing antibodies against CCL17 with validated specificity
Administer competitive inhibitors that displace CCL17 from its receptor
Experimental design considerations:
Include appropriate controls for chemokine redundancy analysis
Perform dose-response studies to identify concentration-dependent effects
Compare responses between wild-type and CCL17-deficient animals in parallel experiments
This multi-faceted approach allows researchers to attribute observed effects specifically to CCL17 activity rather than to other chemokines that may have overlapping functions.
CCL17 provides neuroprotection in subarachnoid hemorrhage (SAH) through a multi-step mechanism:
Primary signaling axis: CCL17 binds to CCR4 receptors on microglia, activating the mTORC2 pathway
Microglial polarization: This activation shifts microglia from pro-inflammatory (M1-like) to anti-inflammatory (M2-like) phenotypes
Morphological changes: Treatment with recombinant CCL17 (rCCL17) reverses SAH-induced alterations in microglial morphology, restoring normal branching patterns, endpoint numbers, and process lengths
Gene expression modification: rCCL17 increases expression of M2-like microglia-associated genes
Neuronal protection: The polarized microglia secrete anti-inflammatory cytokines and growth factors that protect neurons from apoptosis
The neuroprotective effect is abolished by inhibiting either CCR4 (with AZD2098) or mTORC2 (with JR-AB2-011), confirming the specificity of this pathway . In rat SAH models, exogenous rCCL17 administration significantly reduces neuronal death and improves neurological function through this mechanism.
Experimental design for studying CCL17-mediated microglial polarization:
In vivo experimental approach:
Create rat/mouse SAH model using established protocols
Administer rCCL17 intracerebroventricularly at different time points (0, 3, 6, 12, 24 hours post-injury)
Implement the following experimental groups:
Sham (surgery without SAH)
SAH + vehicle (PBS)
SAH + rCCL17
SAH + rCCL17 + CCR4 inhibitor (AZD2098)
SAH + rCCL17 + mTORC2 inhibitor (JR-AB2-011)
Assess microglial morphology using:
Immunofluorescent staining with microglial markers (Iba1)
Morphological analysis with ImageJ plugins to quantify:
Number of branches
Number of endpoints
Process lengths
In vitro validation:
Isolate primary microglia from rat/mouse brains
Stimulate with:
rCCL17 alone
rCCL17 + CCR4 inhibitor
rCCL17 + mTORC2 inhibitor
Analyze polarization markers via:
RT-qPCR for M1/M2 gene expression profiles
Flow cytometry for surface marker expression
Cytokine/chemokine secretion profiles via ELISA/multiplex assays
Key measurements:
mRNA expression of M2-like markers
Protein levels of phosphorylated Akt at Ser473 (mTORC2 activity indicator)
Neuronal apoptosis (TUNEL assay)
Neurological function (behavior tests)
This comprehensive approach enables researchers to establish both the causality and the mechanistic pathway of CCL17's effects on microglial polarization following brain injury.
CCL17 expression in dendritic cells (DCs) demonstrates complex compartmentalized regulation:
Cell type-specific expression:
CCL17 is predominantly expressed in CD11b+ DCs
Activated Langerhans cells show high expression
Mature DCs in lymphoid and non-lymphoid organs express CCL17
Expression is minimal in plasmacytoid DCs
Regulatory mechanisms:
Toll-like receptor (TLR) stimulation: CCL17 expression is upregulated after stimulation with TLR ligands
Cytokine environment: Th2 cytokines (IL-4, IL-13) enhance CCL17 production
Maturation state: Expression increases with DC maturation
Tissue microenvironment: Different expression patterns in skin, lymph nodes, and other tissues
Detection methods:
Method | Application | Sensitivity | Advantages | Limitations |
---|---|---|---|---|
CCL17/EGFP reporter mice | In vivo tracking | High | Cell-specific visualization; real-time monitoring | Requires genetic modification |
RT-qPCR | mRNA quantification | Very high | Precise quantification; specific primers available (e.g., pCCL17-US 5′-CAT GTG AAG AAG GCC ATC AGA TTG GTG-3′ and pCCL17-DS 5′-GAG GGA GGA AGG CTT TAT TCC GTT GC-3′) | No protein-level information |
Flow cytometry | Protein detection | Moderate | Single-cell resolution; phenotypic characterization | Requires cell permeabilization |
ELISA | Secreted protein | Moderate-high | Quantification of secreted protein | No cellular resolution |
Immunohistochemistry | Tissue localization | Moderate | Spatial information; context within tissue | Limited quantification |
Researchers can utilize CCL17/EGFP reporter systems (CCL17E/+ or CCL17E/E) to visualize CCL17-expressing cells in vivo without disrupting tissue architecture , providing valuable insights into the dynamic regulation of CCL17 in different DC populations.
Protocol for studying CCL17-mediated T cell responses:
T cell migration assay:
Isolate CD4+ T cells from lymph nodes and spleen using MACS purification
Pre-activate T cells with anti-CD3/CD28 for 48 hours (to upregulate CCR4)
Prepare chemotaxis chambers:
Add rCCL17 (0.1-100 ng/ml) to lower chamber
Place 5×10⁵ activated T cells in upper chamber
Incubate for 2-3 hours at 37°C
Count migrated cells by flow cytometry
Calculate chemotactic index for each concentration
T cell activation/proliferation assay:
Generate bone marrow-derived or lymph node-derived DCs
Pulse DCs with antigenic peptide (e.g., CRP peptide at 1 μg/ml)
Co-culture 5×10³ DCs with MACS-purified CD4+ T cells
Add rCCL17 at various concentrations (0-100 ng/ml)
Measure proliferation by ³H-thymidine incorporation (add 1 μCi/well during last 17 hours of 72-hour culture)
Alternatively, use CFSE dilution assay to track cell division
Analyze T cell activation markers (CD25, CD69, CD44) by flow cytometry
Measure cytokine production (IL-2, IFN-γ, IL-4) by ELISA
Controls and validation:
Include CCR4 antagonist (AZD2098) to confirm specificity
Compare responses between wild-type and CCR4-deficient T cells
Use CCL17-neutralizing antibodies as additional controls
Include other chemokines (CCL22) that share the CCR4 receptor to assess specificity
This comprehensive approach allows researchers to distinguish CCL17-specific effects on T cell behavior from those mediated by other chemokines or signaling pathways.
Strategic approaches for investigating chemokine redundancy using CCL17 models:
Genetic model utilization:
Compare phenotypes: Analyze CCL17E/E (homozygous knockout) vs. CCL17E/+ (heterozygous) vs. wild-type mice to identify dose-dependent effects
Create compound knockouts: Generate CCL17/CCL22 double knockouts to assess redundancy within the CCR4 ligand family
Cell-specific deletion: Use conditional knockouts (Cre-loxP system) targeting specific cell populations:
DC-specific deletion using CD11c-Cre
Neuronal deletion using Thy1-Cre
Microglial deletion using CX3CR1-CreER
Experimental approaches:
Challenge models: Subject knockout models to various disease challenges:
Neuroinflammation (SAH, stroke models)
Allergic inflammation
Infectious disease models
Compensatory mechanism analysis:
Measure expression of related chemokines (CCL22, CCL1, CCL8) in CCL17-deficient models
Analyze receptor expression changes (CCR4, CCR8) in the absence of CCL17
Profile transcriptome changes in target cells using RNA-seq
Reporter system applications:
Dynamic visualization: Use CCL17/EGFP reporter mice to track spatiotemporal expression patterns in:
Development
Disease progression
Response to therapeutic interventions
Cell isolation strategies: Sort EGFP+ cells from CCL17E/+ mice for detailed molecular profiling
Compound reporter systems: Cross with other reporter strains to simultaneously track multiple components of chemokine networks
This multi-dimensional approach enables researchers to systematically map the redundant and non-redundant functions of CCL17 within complex chemokine networks, revealing both unique and shared roles across different physiological and pathological contexts.
Methodological framework for studying CCL17 signaling in diverse neurological contexts:
Experimental design considerations:
Disease model selection:
Acute models: Traumatic brain injury, ischemic stroke, intracerebral hemorrhage
Chronic models: Alzheimer's disease, Parkinson's disease, multiple sclerosis
Developmental models: Autism spectrum disorders, schizophrenia
Temporal profiling:
Establish baseline CCL17 expression in healthy brain regions
Map temporal expression changes after injury/disease onset (acute, subacute, chronic phases)
Correlate expression with disease progression markers
Cell-specific analysis:
Identify CCL17-producing cells in different neurological conditions
Characterize CCR4 expression on target cell populations
Determine mTORC2 activation status across cell types
Technical approaches:
In vivo signaling visualization:
Implement intravital microscopy with CCL17/EGFP reporter mice
Use phospho-specific antibodies (p-Akt Ser473) to track mTORC2 activation
Apply CLARITY or iDISCO techniques for whole-brain CCL17/CCR4 mapping
Pharmacological interventions:
Design administration protocols for rCCL17 considering blood-brain barrier permeability
Implement CCR4 antagonist (AZD2098) and mTORC2 inhibitor (JR-AB2-011) at disease-appropriate timepoints
Consider combination with standard-of-care treatments
Microglial-specific manipulations:
Outcome measures:
Neurological function assessment (behavior tests specific to each model)
Neuroinflammatory profiling (cytokine/chemokine arrays)
Cellular response characterization (microglial morphology and polarization)
Neurodegeneration/neuroprotection markers (apoptosis, synaptic density)
This comprehensive approach enables researchers to establish whether the CCL17/CCR4/mTORC2 pathway represents a conserved neuroprotective mechanism across different neurological disorders or has context-specific functions that vary by disease etiology and progression.
Common challenges and solutions when working with recombinant CCL17:
Additional technical considerations:
Storage stability:
Species cross-reactivity issues:
Rat CCL17 may have different potency on human vs. rat cells
Validate species-specific activity before cross-species experiments
Consider using species-matched systems when possible
Detection limitations:
Use antibodies validated specifically for rat CCL17
Confirm specificity with appropriate controls (CCL17-deficient samples)
Consider developing sandwich ELISA with capture/detection antibody pairs
These evidence-based solutions help researchers troubleshoot common technical challenges when working with recombinant rat CCL17, ensuring consistent and reliable experimental results.
Strategies for distinguishing endogenous from exogenous CCL17:
Protein modification approaches:
Tagged recombinant proteins:
Fluorescent labeling:
Conjugate recombinant CCL17 with fluorophores (e.g., Alexa Fluor dyes)
Track distribution via microscopy or flow cytometry
Note: Verify that labeling doesn't affect bioactivity
Genetic approaches:
Species differences:
Reporter systems:
Analytical methods:
Temporal discrimination:
Measure baseline CCL17 before exogenous administration
Track concentration changes with high temporal resolution
Account for potential endogenous upregulation due to experimental procedures
Spatial distribution analysis:
Map normal tissue distribution of endogenous CCL17
Identify abnormal distribution patterns after exogenous administration
Use tissue microarrays for high-throughput analysis
Mass spectrometry approaches:
Use isotope-labeled recombinant CCL17 for exogenous administration
Perform mass spectrometry to differentiate labeled vs. unlabeled protein
Enables precise quantification of both pools simultaneously
This comprehensive toolkit allows researchers to accurately track and distinguish exogenous CCL17 from endogenous production, enabling more precise interpretation of experimental results in complex biological systems.