Immune Cell Recruitment: Chemoattracts dendritic cells, T cells (Th17, Tregs), B cells, and Langerhans cells to sites of inflammation .
Inflammatory Regulation: Upregulated by TNF-α, IL-17, and LPS, but suppressed by IL-10 and IFN-γ .
Antimicrobial Activity: Exhibits antibacterial effects against E. coli and S. aureus .
Ccl20 and CCR6 are significantly upregulated in psoriatic skin lesions (7-fold and 4-fold, respectively) compared to healthy tissue. This drives the migration of CLA+ T cells to inflamed epidermis .
Neutralizing Ccl20 with monoclonal antibodies blocks calcium mobilization in CCR6-expressing cells .
Regional neural activation (e.g., sensory nerve stimulation) induces Ccl20 expression via IL-6 amplifier signaling, facilitating pathogenic CD4+ T cell entry into the CNS in autoimmune encephalitis models .
Ccl20 enhances sperm motility and chemotaxis via CCR6-mediated calcium signaling, highlighting its role in reproductive physiology .
Chemotaxis Assays: Used to study immune cell migration (e.g., Transwell assays) .
Calcium Mobilization: Monitored via Indo-1A loading in CCR6-transfected cells .
Recombinant mouse CCL20 is a low molecular weight chemokine (approximately 8.0 kDa) expressed as a tag-free protein spanning amino acids 28-97 of the native sequence. The protein contains the characteristic CC chemokine fold but exhibits unusual properties compared to other CC chemokines. Unlike most CC chemokines, crystal structures of CCL20 (PDB codes 2HCI and 1M8A) reveal that it can adopt a CXC-type dimeric arrangement, although NMR studies indicate this self-association is relatively weak and pH dependent . The mature recombinant protein produced in E. coli expression systems contains the full biological activity domain without additional tags that might interfere with receptor binding or structural studies .
Mouse CCL20 interacts specifically with the G-protein coupled receptor CCR6. This exclusive relationship between CCL20 and CCR6 is relatively unique in the chemokine family, as most chemokines can bind multiple receptors. The CCL20-CCR6 interaction mediates several critical biological processes including:
Chemoattraction of dendritic cells and T lymphocytes (particularly Th17 cells)
Recruitment of immune cells to mucosal surfaces
Regulation of inflammatory responses
The binding of CCL20 to CCR6 triggers various signaling cascades that lead to cell migration, calcium mobilization, and gene expression changes. The specificity of this interaction makes it a potential target for therapeutic interventions in inflammatory conditions .
Researchers can distinguish recombinant mouse CCL20 from other chemokines through several approaches:
Receptor binding specificity: Using CCR6-expressing cells in competitive binding assays
Antibody-based detection: Employing specific antibodies like the mouse CCL20/MIP-3 alpha Antibody (Clone #114908) in Western blotting, ELISA, or immunohistochemistry
Functional assays: Measuring chemotaxis of CCR6+ cells (e.g., BaF3 cells transfected with human CCR6)
Mass spectrometry: Confirming molecular weight (8.0 kDa for tag-free mouse CCL20)
SDS-PAGE mobility: Monitoring migration patterns under reducing and non-reducing conditions
For optimal specificity, researchers should perform neutralization experiments using CCL20-specific antibodies to confirm that observed effects are truly CCL20-dependent. For example, chemotaxis elicited by recombinant mouse CCL20 (20 ng/mL) can be neutralized by increasing concentrations of anti-mouse CCL20 monoclonal antibody, with an ND50 typically between 3-15 μg/mL .
For optimal stability and biological activity, recombinant mouse CCL20 should be handled as follows:
Reconstitution Protocol:
Briefly centrifuge the lyophilized protein vial to collect all material at the bottom
Reconstitute in sterile PBS containing at least 0.1% carrier protein (BSA or HSA)
Gently mix by pipetting or brief vortexing, avoiding foam formation
Allow the protein to sit at room temperature for 10-15 minutes before use
Storage Conditions:
Short-term (≤1 month): Store at 4°C in sterile conditions
Long-term: Store at -20°C to -80°C in working aliquots to avoid repeated freeze-thaw cycles
Add a preservative (e.g., 0.1% sodium azide) for longer storage periods when stored at 4°C
Monitor activity periodically through functional assays to ensure protein viability
Researchers should validate protein activity after reconstitution using a chemotaxis assay with CCR6-expressing cells before employing the protein in critical experiments .
Evaluating the biological activity of recombinant mouse CCL20 requires multiple complementary approaches:
Chemotaxis Assay:
The most direct functional assessment involves measuring CCL20-induced migration of CCR6-expressing cells. The BaF3 mouse pro-B cell line transfected with human CCR6 is commonly used for this purpose. In a dose-dependent chemotaxis assay, recombinant mouse CCL20 should induce cell migration at concentrations as low as 1-10 ng/mL, with optimal activity typically observed at 20-50 ng/mL. Cell migration can be quantified using Resazurin or similar viability dyes to measure the number of cells that migrate through a membrane .
Calcium Mobilization Assay:
Binding of CCL20 to CCR6 triggers intracellular calcium flux, which can be measured using calcium-sensitive fluorescent dyes like Fluo-4 AM. Researchers should observe a rapid and transient increase in fluorescence intensity following CCL20 stimulation.
Receptor Binding Assay:
Competitive binding assays using radiolabeled or fluorescently labeled CCL20 can determine binding affinity. The IC50 value for unlabeled CCL20 displacing labeled CCL20 provides an indirect measure of biological activity.
Cell Signaling Assays:
Western blotting for phosphorylated ERK1/2, Akt, or other downstream signaling molecules can confirm functional receptor activation. Researchers should observe time-dependent and dose-dependent changes in phosphorylation status following CCL20 stimulation.
To ensure specificity, control experiments should include:
CCR6-negative cells (negative control)
Anti-CCL20 neutralizing antibodies
CCR6 receptor antagonists
Heat-inactivated CCL20 (denatured control)
Engineering modified forms of mouse CCL20 can yield variants with altered receptor binding, activation properties, or stability. Key approaches include:
Site-Directed Mutagenesis:
Strategic amino acid substitutions can create CCL20 variants with modified properties. For example, the S64C variant creates a constitutive dimer through introduction of a cysteine that forms an intermolecular disulfide bond. This engineered dimer binds and partially activates CCR6 but inhibits T cell chemotaxis, demonstrating potential therapeutic value .
The engineering process typically involves:
Structure-based design using algorithms like Disulfide by Design
Visual inspection of crystal structures (such as PDB ID 2HCI) to identify suitable modification sites
Creating a panel of variants (e.g., V21C/T24C, G22C/T24C, F23C, V60C/V67C, S64C)
Expression in E. coli and refolding
Biophysical characterization to confirm proper folding and oligomeric state
Functional testing to determine altered properties
Domain Swapping:
Chimeric proteins created by exchanging domains between CCL20 and other chemokines can yield insights into structure-function relationships and potentially create novel activities.
Fusion Proteins:
Linking CCL20 to other proteins can create bifunctional molecules with expanded capabilities, such as:
CCL20-Fc fusion for extended half-life
CCL20-toxin conjugates for targeted cell killing
CCL20-reporter fusions for tracking receptor binding
Post-Translational Modifications:
Introduction of specific glycosylation sites or other modifications can alter stability, receptor binding, or immunogenicity.
Researchers should confirm proper folding of engineered variants using techniques such as circular dichroism, nuclear magnetic resonance (NMR) spectroscopy, or X-ray crystallography .
CCL20 plays a crucial role in the pathogenesis of psoriasis through several mechanisms:
T Cell Recruitment: CCL20 is upregulated in psoriatic lesions and attracts CCR6-expressing Th17 cells to the skin, which are key drivers of psoriatic inflammation
Dendritic Cell Trafficking: CCL20 mediates recruitment of CCR6+ dendritic cells that present antigens and stimulate T cell activation
Inflammatory Amplification Loop: In response to skin injury, keratinocytes produce CCL20, which recruits immune cells that release pro-inflammatory cytokines (IL-17, IL-22, IL-23), further stimulating CCL20 production
Epithelial Barrier Disruption: CCL20-recruited inflammatory cells release cytokines that alter keratinocyte differentiation, contributing to the characteristic epidermal hyperplasia
In mouse models of psoriasis, such as the IL-23-dependent model, engineered CCL20 variants that bind CCR6 but inhibit T cell chemotaxis have been shown to reduce disease severity. The disulfide-linked CCL20 dimer (S64C variant) demonstrates therapeutic potential by blocking the recruitment of pathogenic T cells to the skin while partially activating the CCR6 receptor .
This suggests that targeting the CCL20-CCR6 axis represents a promising approach for treating psoriasis and potentially other inflammatory skin conditions.
CCL20 contributes to cancer progression through multiple mechanisms:
Cancer-Promoting Mechanisms of CCL20:
Immunosuppression: CCL20 can recruit regulatory T cells (Tregs) to the tumor microenvironment, suppressing anti-tumor immune responses
Angiogenesis: CCL20 promotes formation of new blood vessels to support tumor growth
Epithelial-to-Mesenchymal Transition (EMT): CCL20 signaling induces EMT, enhancing cancer cell invasiveness
Migration/Invasion: Activation of CCR6 by CCL20 stimulates cancer cell motility through various signaling pathways
Chemoresistance: CCL20 can induce resistance to chemotherapeutic agents through activation of survival pathways
CCL20 is particularly implicated in breast cancer progression, where it has emerged as a potential therapeutic target .
Therapeutic Targeting Approaches:
Monoclonal Antibodies: Neutralizing antibodies against CCL20 can block its interaction with CCR6
Receptor Antagonists: Small molecule inhibitors of CCR6 can prevent CCL20-mediated signaling
Engineered CCL20 Variants: Modified versions of CCL20 that bind CCR6 without activating downstream signaling can serve as competitive inhibitors
Signaling Pathway Inhibitors: Targeting the downstream pathways activated by CCL20/CCR6 interaction
Combination Therapies: Blocking CCL20 in combination with immune checkpoint inhibitors may enhance anti-tumor immunity
For effective therapeutic development, researchers must consider the complex cytokine network within the tumor microenvironment, as multiple factors collectively regulate tumor progression. Understanding the signaling pathways that regulate CCL20 function provides additional targets for intervention .
Several experimental models are valuable for investigating CCL20 function in immune regulation:
In Vitro Models:
Chemotaxis Assays: Using transwell migration systems with CCR6-expressing cells (e.g., BaF3 cells transfected with CCR6, Th17 cells, or dendritic cells) to assess CCL20-mediated cell recruitment
3D Organoid Cultures: Intestinal or skin organoids can recapitulate tissue architecture and demonstrate CCL20's role in immune cell recruitment to epithelial surfaces
Co-culture Systems: Combining epithelial cells with immune cells to study CCL20-mediated interactions
In Vivo Models:
IL-23-Induced Psoriasis Model: Intradermal injection of IL-23 induces psoriasiform skin inflammation dependent on CCL20-CCR6 interaction, allowing assessment of therapeutic interventions targeting this axis
Imiquimod-Induced Psoriasis Model: Topical application of imiquimod cream induces psoriasis-like inflammation with upregulation of CCL20
DSS-Induced Colitis: Dextran sodium sulfate administration causes intestinal inflammation where CCL20 plays a role in immune cell recruitment
CCR6 and CCL20 Knockout Mice: Genetic models lacking CCR6 or CCL20 help elucidate their roles in homeostasis and inflammation
Humanized Mouse Models: Mice reconstituted with human immune cells allow study of human CCL20-CCR6 interactions
Experimental Readouts:
Flow cytometry for immune cell recruitment and phenotyping
Histological assessment of tissue inflammation
Cytokine profiling by ELISA or multiplex assays
Gene expression analysis by qPCR or RNA-seq
In vivo imaging of fluorescently labeled immune cells
The choice of model should be guided by the specific research question, with consideration of species differences in CCL20 function between mice and humans .
Accurate quantification of CCL20 expression requires selecting appropriate methods based on the experimental context:
Protein Level Quantification:
ELISA (Enzyme-Linked Immunosorbent Assay):
Sandwich ELISA using capture and detection antibodies specific for mouse CCL20
Typical detection range: 15-1000 pg/mL
Sample types: Cell culture supernatants, tissue lysates, serum, plasma
Note: For optimal sensitivity, use antibody pairs validated for mouse CCL20 detection
Western Blotting:
Use of specific anti-CCL20 antibodies (e.g., Clone #114908)
Sample preparation: Reduce samples with DTT or β-mercaptoethanol
Expected molecular weight: ~8.0 kDa
Controls: Recombinant mouse CCL20 as positive control
Luminex/Multiplex Assays:
Allow simultaneous detection of CCL20 alongside other cytokines
Useful for comprehensive analysis of inflammatory profiles
Higher throughput than traditional ELISA
Mass Spectrometry:
For unbiased detection and absolute quantification
Particularly useful in complex biological samples
Requires appropriate sample preparation and internal standards
mRNA Level Quantification:
Quantitative Real-Time PCR (qRT-PCR):
Design primers specific for mouse Ccl20 gene
Normalize to appropriate housekeeping genes (e.g., GAPDH, β-actin)
Include no-template and no-RT controls
RNA-Sequencing:
Provides comprehensive transcriptome analysis
Allows detection of alternative splicing variants
Requires bioinformatic analysis pipelines
In Situ Detection:
Immunohistochemistry/Immunofluorescence:
Use of validated anti-CCL20 antibodies on tissue sections
Include appropriate positive and negative controls
Consider co-staining for cell type markers to identify CCL20-producing cells
RNA in situ Hybridization:
Detection of Ccl20 mRNA in tissue sections
Useful for localizing expression at cellular level
RNAscope or similar technologies provide single-molecule sensitivity
When comparing CCL20 levels across different experimental conditions, researchers should maintain consistent sample collection, processing, and analysis methods to minimize technical variability .
Producing high-quality recombinant mouse CCL20 presents several challenges, each with specific solutions:
Challenges and Solutions in Production:
Challenge | Solution | Technical Details |
---|---|---|
Maintaining proper folding | Optimized refolding protocols | Use of redox buffer systems (reduced/oxidized glutathione) to promote correct disulfide bond formation; gradual dialysis to remove denaturants |
Achieving high purity | Multi-step purification strategy | Combination of ion exchange chromatography, size exclusion chromatography, and reverse-phase HPLC; purity should exceed 95% as determined by SDS-PAGE |
Endotoxin contamination | Endotoxin removal | Polymyxin B columns or Triton X-114 phase separation; validate with LAL assay; maintain levels <1.0 EU/μg protein |
Protein aggregation | Stabilizing formulation | Include carrier proteins (e.g., 0.1% BSA); optimize pH and salt concentration; consider addition of non-ionic detergents |
Verification of biological activity | Functional testing | Chemotaxis assays using CCR6+ cells; calcium mobilization assays; receptor binding assays |
Post-translational modifications | E. coli vs. eukaryotic expression | Use of E. coli for tag-free protein; mammalian cells when glycosylation is required |
Expression Systems Comparison:
E. coli Expression System:
Advantages: High yield, cost-effective, tag-free expression possible
Disadvantages: Lacks post-translational modifications, inclusion body formation common
Best practices: Expression as inclusion bodies followed by denaturation and refolding
Mammalian Expression System:
Advantages: Proper post-translational modifications, direct secretion into medium
Disadvantages: Lower yield, higher cost, potential for heterogeneous glycosylation
Best practices: Use of optimized signal sequences, serum-free adaptation
Insect Cell Expression System:
Advantages: Higher yield than mammalian cells, some post-translational modifications
Disadvantages: Different glycosylation patterns than mammalian systems
Best practices: Optimization of multiplicity of infection and harvest time
The most commonly used system for recombinant mouse CCL20 production is E. coli, as evidenced by commercially available products. For optimal quality, purified protein should be subjected to rigorous quality control testing, including mass spectrometry, SDS-PAGE under reducing and non-reducing conditions, and functional assays to confirm biological activity .
Structural analysis of CCL20 provides critical insights for rational drug design of therapeutic antagonists or agonists:
Key Structural Features Informing Drug Design:
Dimerization Interface: The unusual CXC-type dimeric arrangement observed in CCL20 crystal structures (PDB ID codes 2HCI and 1M8A) presents opportunities for designing molecules that can stabilize or disrupt dimerization. The successful engineering of the S64C variant demonstrates how introducing an intermolecular disulfide bond can create a stable dimer with altered function, inhibiting T cell chemotaxis while maintaining receptor binding .
Receptor Binding Domains: Structural studies identifying the precise regions of CCL20 that interact with CCR6 can inform the design of:
Peptide mimetics that compete for receptor binding
Small molecules that disrupt the protein-protein interaction
Antibodies targeting specific epitopes involved in receptor recognition
Conformational Dynamics: NMR studies indicating that CCL20 self-association is weak and pH-dependent suggest that pH-sensitive variants could be developed for targeted activity in specific tissue microenvironments.
Structure-Based Drug Design Approaches:
Computational Modeling:
Molecular dynamics simulations to identify stable binding conformations
Virtual screening of compound libraries against CCL20 or CCR6 binding pockets
In silico prediction of binding affinities for candidate molecules
Biophysical Characterization:
X-ray crystallography of CCL20 complexed with receptor fragments
NMR studies to map binding interfaces
Surface plasmon resonance to determine binding kinetics
Rational Protein Engineering:
Structure-guided mutations to create variants with enhanced receptor affinity but reduced signaling capacity (antagonists)
Engineering of constitutive dimers like CCL20 S64C that demonstrate partial receptor activation
Creation of fusion proteins that combine CCL20 with other functional domains
Therapeutic Applications Based on Structure:
The engineered CCL20 S64C dimer has already demonstrated therapeutic potential in a mouse model of psoriasis, suggesting broader applications for engineered CCL20 variants in:
Treatment of autoimmune diseases (rheumatoid arthritis, multiple sclerosis)
Cancer immunotherapy
Modulation of mucosal immunity
Treatment of inflammatory bowel diseases
By combining structural insights with functional studies, researchers can develop more potent and selective CCL20 inhibitors or modulators with improved pharmacokinetic properties and reduced off-target effects .
The CCL20-CCR6 interaction activates multiple signaling pathways that orchestrate diverse cellular responses:
Primary Signaling Cascades:
G Protein-Coupled Signaling:
Gαi protein activation leads to inhibition of adenylyl cyclase and reduction in cAMP levels
Gβγ subunits activate phospholipase C (PLC), leading to formation of IP3 and DAG
IP3 triggers calcium release from intracellular stores, important for cellular motility
DAG activates protein kinase C (PKC), regulating cytoskeletal reorganization
MAPK Pathways:
ERK1/2 activation promotes cell proliferation and survival
p38 MAPK regulates inflammatory cytokine production
JNK pathway activation influences cell migration and stress responses
PI3K/Akt Pathway:
Phosphatidylinositol 3-kinase (PI3K) activation leads to Akt phosphorylation
Promotes cell survival, metabolism, and cytoskeletal rearrangements
Regulates mammalian target of rapamycin (mTOR) signaling
Small GTPases:
Activation of Rho family GTPases (RhoA, Rac1, Cdc42)
Critical for actin polymerization, cell polarization, and directional migration
Controls formation of lamellipodia and filopodia during chemotaxis
Cell Type-Specific Responses:
Cell Type | Predominant Signaling | Major Functional Outcomes |
---|---|---|
Dendritic cells | PI3K/Akt, Rac1 | Migration, maturation, antigen presentation |
T lymphocytes (Th17) | MAPK, calcium mobilization | Chemotaxis, cytokine production, proliferation |
Epithelial cells | NF-κB, STAT3 | Inflammatory mediator production, barrier function |
Cancer cells | PI3K/Akt, ERK1/2 | EMT, invasion, survival, chemoresistance |
Regulatory Mechanisms:
Receptor Desensitization:
GRK-mediated phosphorylation of CCR6
β-arrestin recruitment leading to receptor internalization
Temporal limitation of signaling
Crosstalk with Other Pathways:
Integration with TLR signaling in dendritic cells
Synergy with cytokine receptor signaling (e.g., IL-17R)
Modulation by microenvironmental factors
Understanding these signaling pathways provides opportunities for therapeutic targeting, either at the level of CCL20-CCR6 interaction or at downstream signaling nodes. Pathway-specific inhibitors could potentially modulate specific aspects of CCL20 function while preserving others, allowing for more precise therapeutic interventions in diseases where CCL20 plays a pathogenic role .
Researchers frequently encounter several challenges when working with CCL20 in experimental settings. Here are common issues and troubleshooting approaches:
Problem | Potential Causes | Troubleshooting Approach |
---|---|---|
Low or variable cell migration | Degraded CCL20 protein | Use freshly reconstituted protein; add protease inhibitors |
Receptor desensitization | Rest cells in cytokine-free media before assay | |
Improper concentration gradient | Optimize CCL20 concentration range (typically 10-100 ng/mL) | |
Variable CCR6 expression | Verify receptor expression by flow cytometry | |
Membrane pore size issues | Adjust pore size based on cell type (5-8 μm optimal) |
Problem | Potential Causes | Troubleshooting Approach |
---|---|---|
Loss of biological activity | Improper reconstitution | Use carrier protein (0.1% BSA) in reconstitution buffer |
Oxidation of methionine residues | Add reducing agents; limit exposure to oxidizing conditions | |
Adsorption to surfaces | Use low-binding tubes; include carrier proteins | |
Repeated freeze-thaw cycles | Aliquot protein before freezing; avoid multiple freeze-thaws | |
Bacterial contamination | Filter solutions; add preservatives for long-term storage |
Problem | Potential Causes | Troubleshooting Approach |
---|---|---|
Non-specific detection | Cross-reactivity with related chemokines | Use monoclonal antibodies with validated specificity |
High background in immunostaining | Optimize blocking conditions; include FcR blocking | |
Non-specific Western blot bands | Verify specificity using recombinant protein control | |
Variable ELISA results | Run standard curve with each assay; use certified antibody pairs |
Problem | Potential Causes | Troubleshooting Approach |
---|---|---|
Low or undetectable Ccl20 mRNA | Timing of sample collection | CCL20 expression can be transient; optimize collection timing |
RNA degradation | Use RNase inhibitors; verify RNA integrity | |
Suboptimal primer design | Design primers spanning exon-exon junctions | |
Inadequate stimulation | Verify stimulation protocol with positive controls (e.g., TNF-α, IL-1β) |
Problem | Potential Causes | Troubleshooting Approach |
---|---|---|
Poor protein folding | Disruption of critical residues | Use structure-guided mutagenesis; avoid conserved regions |
Inefficient disulfide formation | Optimize redox conditions during refolding | |
Aggregation during refolding | Use pulsatile dilution method; add stabilizing agents | |
Improper purification | Multi-step purification strategy; verify by mass spectrometry |
When troubleshooting, researchers should systematically test each variable while maintaining appropriate positive and negative controls. For example, when evaluating chemotaxis, include a known chemoattractant as positive control and buffer-only conditions as negative control .
The tumor microenvironment (TME) profoundly influences CCL20 expression and function through multiple regulatory mechanisms:
Regulatory Factors in the Tumor Microenvironment:
Hypoxia:
Hypoxic conditions activate HIF-1α, which can upregulate CCL20 expression
Creates a feedback loop enhancing angiogenesis and immune cell recruitment
Cancer cells in hypoxic regions produce more CCL20, promoting invasion and metastasis
Inflammatory Cytokines:
TNF-α, IL-1β, and IL-17 strongly induce CCL20 expression in tumor and stromal cells
NF-κB pathway activation serves as a major mediator of cytokine-induced CCL20 expression
Cytokine-rich environments amplify CCL20 production, creating chemotactic gradients
Tumor-Associated Macrophages (TAMs):
M2-polarized TAMs can produce CCL20, recruiting CCR6+ regulatory T cells
This creates an immunosuppressive microenvironment favoring tumor growth
TAM-derived factors may also enhance tumor cell expression of CCL20
Microbial Products:
In certain cancers (e.g., colorectal cancer), microbial components like LPS stimulate CCL20 production
Pattern recognition receptor activation links microbiome alterations to CCL20-mediated inflammation
Signaling Pathways Regulating CCL20 in Cancer:
NF-κB Pathway:
Primary transcriptional regulator of CCL20 expression
Activated by inflammatory cytokines, growth factors, and cellular stress
Inhibition of NF-κB can substantially reduce CCL20 production
STAT3 Signaling:
Activated by IL-6 family cytokines abundant in the TME
Promotes CCL20 expression and CCR6 upregulation on cancer cells
Creates autocrine/paracrine signaling loops enhancing tumor progression
Wnt/β-catenin Pathway:
Dysregulated in many cancers and can promote CCL20 expression
Links developmental pathways to chemokine production
β-catenin accumulation correlates with increased CCL20 levels in some tumors
MAPK Pathways:
ERK1/2 activation enhances CCL20 production in response to growth factors
p38 MAPK mediates stress-induced CCL20 expression
JNK pathway regulates CCL20 in response to inflammatory stimuli
Functional Consequences in Cancer Progression:
CCL20 produced in the TME contributes to cancer progression through:
Immunosuppression: Recruitment of CCR6+ regulatory T cells that suppress anti-tumor immunity
Angiogenesis: Stimulation of endothelial cell migration and tube formation
EMT Promotion: Induction of epithelial-to-mesenchymal transition in cancer cells
Invasion/Metastasis: Enhanced migration and matrix degradation capacity
Chemoresistance: Activation of survival pathways protecting against apoptosis
Understanding these complex regulatory networks provides opportunities for therapeutic targeting of CCL20 in cancer, potentially through: