Fact-checked by Grok 2 weeks ago

CCL20

CCL20, also known as C-C motif chemokine ligand 20 (or macrophage inflammatory protein-3 alpha, MIP-3α), is a small secreted cytokine belonging to the CC subfamily of chemokines that plays a crucial role in directing the migration of immune cells, particularly by acting as a chemoattractant for lymphocytes (especially memory T cells and Th17 cells), immature dendritic cells, and B cells through its specific interaction with the G protein-coupled receptor CCR6. Encoded by the CCL20 gene (formerly SCYA20) located on the long arm of human chromosome 2 at position 2q36.3, the protein is synthesized as a 95- or 96-amino-acid precursor with a 21- or 22-amino-acid signal peptide, yielding a mature form of approximately 70 amino acids and a molecular weight of about 8 kDa; it features a characteristic CC motif where two adjacent cysteine residues are involved in disulfide bond formation critical for its structure and function. CCL20 was independently cloned and characterized in 1997 by multiple research groups using bioinformatics and expression screening approaches, with initial reports identifying it as "Exodus-1" or MIP-3α due to its expression patterns in lymphoid tissues and its role in attracting immune cells from peripheral sites to mucosal and inflamed areas. The is constitutively expressed at low levels in tissues such as the liver, , and mucosal epithelia (e.g., in the intestines and ), but its production is strongly upregulated by proinflammatory stimuli including tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and (LPS), enabling rapid recruitment of CCR6-expressing cells to sites of infection or injury. In terms of biological function, CCL20-CCR6 signaling is essential for mucosal immunity and , facilitating the trafficking of regulatory T cells (Tregs) and effector T cells to barrier sites like the gut and , where it helps balance tolerance and inflammation; however, dysregulation of this axis contributes to various pathologies. Notably, elevated CCL20 expression is implicated in autoimmune and inflammatory diseases, including (where it drives Th17 cell infiltration into the ), inflammatory bowel disease (IBD, via promotion of pathogenic T cell homing to the gut), , , and certain cancers such as colorectal carcinoma, where it supports tumor progression through autocrine/paracrine mechanisms. Structurally, CCL20 adopts a monomeric or dimeric form with a disordered that engages the CCR6 receptor in a shallow binding pocket, triggering activation and downstream signaling for , as revealed by cryo-electron microscopy studies. Therapeutically, the CCL20-CCR6 pathway has emerged as a promising target for , with experimental antagonists and neutralizing antibodies showing efficacy in preclinical models of and IBD by blocking leukocyte recruitment and reducing ; as of 2025, novel orally available CCR6 inhibitors like IDOR-1117-2520 and structural insights into allosteric antagonists have further advanced potential treatments.

Discovery and Nomenclature

Discovery

CCL20, a member of the CC family, was first identified in 1997 through independent cloning efforts by multiple research groups. Hieshima et al. identified partial cDNA sequences encoding CCL20 using a PCR-based signal trap method from cDNA libraries including fetal and HepG2 hepatoma cells, designating it as liver and activation-regulated (LARC) due to its expression in liver and upregulation upon activation. Concurrently, Hromas et al. cloned the human and murine homologs from a pancreatic cDNA library, naming it based on its selective expression in lymphoid tissues and chemotactic properties. In parallel, bioinformatics approaches led to its discovery as macrophage inflammatory protein-3α (MIP-3α). Rossi et al. identified the sequence from databases, predicting its role as a proinflammatory based on structural to other chemokines. These initial clonings revealed a 70-amino-acid mature protein with a characteristic motif and demonstrated constitutive expression in liver and , with inducible expression in response to proinflammatory stimuli such as TNF-α and . Shortly thereafter, CCL20 was characterized as the specific for the CCR6 (initially termed STRL22). et al. reported high-affinity binding of MIP-3α to STRL22-transfected cells, inducing calcium mobilization and , establishing CCL20 as its sole known ligand. Early functional studies confirmed its potent chemoattractant activity for memory T lymphocytes and immature dendritic cells, highlighting its role in directing immune cell trafficking to mucosal and inflamed sites.

Nomenclature

The official for CCL20, as designated by the International Union of Basic and Clinical Pharmacology (IUPHAR), is C-C ligand 20. This systematic name reflects its classification within the CC subfamily, characterized by the conserved cysteine , and its sequential numbering among known ligands. CCL20 has several alternative names stemming from its independent discoveries in 1997. It was initially termed macrophage inflammatory protein 3-alpha (MIP-3α) due to its proinflammatory properties and structural similarity to other macrophage inflammatory proteins like MIP-1α. Another name, liver and -regulated (LARC), originated from observations of its constitutive expression in the liver and upon T-cell . Additional synonyms include small-inducible A20 (SCYA20), reflecting its role as a small secreted , and beta- -1 (or simply ), named for its expression in lymphoid tissues such as the and lymph nodes, evoking the "exodus" of lymphocytes. These names are used interchangeably in , with CCL20 now the preferred standard. The human gene symbol is , located on , while orthologs in other species include .

Genomics

Gene Location and Organization

The gene is located on the long (q) arm of human at the cytogenetic band 2q36.3. Its genomic coordinates on the GRCh38 assembly span from 227,813,842 to 227,817,556, encompassing approximately 3.7 kb of DNA on the forward strand. The is organized into 4 exons interrupted by 3 introns, with the exon-intron boundaries conforming to the GT-AG rule typical of eukaryotic . Two transcript variants arise from : the longer isoform 1 (NM_004591.3, 291 coding sequence) and the shorter isoform 2 (NM_001130046.2), which utilizes an alternate splice site in the 3' region but preserves the , resulting in a protein of similar length; no major functional isoforms with distinct roles have been reported. The promoter region upstream of the transcription start site includes binding sites for key transcription factors, such as (located approximately -80 to -70 bp) and AP-1, which facilitate inflammatory regulation of CCL20 expression.

Sequence Features

The CCL20 gene's cDNA contains an of 288 base pairs that encodes a 96-amino-acid precursor protein. This precursor consists of an N-terminal of 26 amino acids followed by the mature protein of 70 amino acids. The is cleaved upon , yielding the functional mature form. The sequence of mature CCL20 begins with Ala-Ala-Ser-Asn-Phe-Asp-Cys-Cys-Leu-Gly-... and ends with Lys-Lys-Val-Lys-Asn-Met. It features the conserved motif at positions 7 and 8 (Cys7-Cys8), which forms intramolecular bonds essential for structural stability—specifically, Cys7-Cys34 and Cys8-Cys50. Unlike ELR+ CXC that possess an N-terminal Glu-Leu-Arg (ELR) motif promoting , CCL20 lacks this motif, aligning with its role in immune cell recruitment rather than vascular effects. Mature CCL20 exhibits 20-30% sequence identity to other chemokines, including CCL19 and CCL21, underscoring shared family characteristics such as the motif and bonding pattern while distinguishing its unique receptor specificity for CCR6.

Protein Structure

Primary and Secondary Structure

The CCL20 encodes a precursor protein of 96 , which includes a hydrophobic N-terminal of 26 residues that directs the protein to the secretory pathway. Upon cleavage of this between Ala26 and Asn27, the mature CCL20 protein is produced, consisting of 70 with a calculated of approximately 8 kDa. The primary sequence of the mature form is Asn-Phe-Asp-Cys-Cys-Leu-Gly-Tyr-Thr-Asp-Arg-Ile-Leu-His-Pro-Lys-Phe-Ile-Val-Gly-Phe-Thr-Arg-Gln-Leu-Ala-Asn-Glu-Gly-Cys-Asp-Ile-Asn-Ala-Ile-Ile-Phe-His-Thr-Lys-Lys-Lys-Leu-Ser-Val-Cys-Ala-Asn-Pro-Lys-Gln-Thr-Trp-Val-Lys-Tyr-Ile-Val-Arg-Leu-Leu-Ser-Lys-Lys-Val-Lys-Asn-Met. The secondary structure of mature CCL20 adopts the canonical chemokine fold, characterized by an N-terminal domain forming three antiparallel β-strands (β1: residues 20–26, β2: 36–41, β3: 46–49) and a C-terminal α-helix (residues 54–63) that packs against the β-sheet core. This arrangement is typical of the chemokine subfamily, where the flexible N-terminal loop preceding the β1 strand contributes to receptor specificity. The structural integrity of CCL20 is maintained by two conserved intramolecular disulfide bridges involving its four cysteine residues, linking Cys4 to Cys30 and Cys5 to Cys46 (numbered from the ); these bonds rigidly connect the adjacent cysteines of the CC to the subsequent cysteines in the β1 and β3 strands, essential for the overall fold and stability. Post-translational modifications of CCL20 are minimal, with no N-linked sites present in the due to the absence of the Asn-X-Ser/Thr ; the protein is secreted predominantly in its unmodified, non-glycosylated form to facilitate rapid immune cell recruitment.

Tertiary Structure and Function

The tertiary of CCL20 features a conserved fold consisting of a central core of three antiparallel β-strands (β1: residues 20-26, β2: 36-41, β3: 46-49) packed against an overlying C-terminal α- (residues 54-63), stabilized by two intramolecular bonds between Cys4-Cys30 and Cys5-Cys46. This architecture is evident in the crystal of human CCL20 (PDB : 1M8A), resolved at 1.7 Å, which reveals a disordered (residues 1-3) preceding an N-loop (residues 4-19) and a short 3₁₀- (residues 15-17). The NMR of human CCL20 (PDB : 2JYO) confirms this monomeric fold under acidic conditions, with the β-sheet forming a hydrophobic core that supports the . CCL20 exhibits dimerization in solution and crystal structures, adopting an atypical α-helical dimer for a CC , where the C-terminal helices from two monomers pack together, burying approximately 1155 Ų of surface area and forming a six-stranded antiparallel β-sheet through β1-strand interactions. This dimeric state is captured in the (PDB : 1M8A) and a more recent high-resolution structure (PDB : 7T1E) at 1.46 Å, which also includes a monomeric form alongside two dimers. Dimerization is -dependent, favoring dimers at neutral due to the protonation state of His14 and His38, while shifting to monomers at lower (e.g., pH 5.4), as determined by NMR diffusion and titration studies; this equilibrium influences chemotactic potency, with dimers showing reduced activity compared to monomers in receptor activation assays. Key functional domains include the N-loop, which facilitates initial docking to the CCR6 receptor by interacting with extracellular loops (particularly ECL3), enabling subsequent N-terminal insertion for signaling initiation. The C-terminal α-helix, rich in basic residues (e.g., Lys55, Arg59, Lys66), mediates binding to glycosaminoglycans (GAGs) such as heparan sulfate, promoting immobilization on extracellular matrices and haptotactic gradients for immune cell recruitment. These structural elements underscore CCL20's role in targeted chemotaxis, with the dimer's altered conformation potentially modulating GAG affinity and receptor engagement efficiency.

Expression and Regulation

Tissue and Cellular Expression

CCL20 exhibits basal expression predominantly in mucosal tissues, where it plays a key role in immune surveillance at barrier sites. High levels are observed in the intestines, particularly by intestinal epithelial cells, as well as in the lungs via pulmonary epithelial cells and in the through . Additionally, constitutive expression occurs in the liver and , contributing to steady-state immune in these organs. At the cellular level, CCL20 is primarily produced by epithelial cells in a constitutive manner, but it can also be induced in endothelial cells and fibroblasts in response to inflammatory cues. Expression becomes inducible in immune cells such as macrophages and T cells, particularly Th17 subsets, in response to inflammatory cues, allowing for dynamic regulation during immune activation. During embryonic development, CCL20 expression is upregulated in lymphoid tissues, coinciding with the of secondary lymphoid structures. In the intestine, CCL20 mRNA appears in as early as embryonic day 17.5, driven by lymphotoxin signaling, which supports the of lymphoid tissue inducer cells. This pattern facilitates the formation of mucosal lymphoid aggregates essential for postnatal immunity. The expression patterns of CCL20 are well-conserved across , with similar and cellular distributions observed in humans and mice. Both exhibit high basal levels in mucosal epithelia and lymphoid organs, as evidenced by comparable promoter structures and inducible responses in immune cells.

Regulatory Mechanisms

The expression of the CCL20 gene is primarily regulated at the transcriptional level by key transcription factors responsive to proinflammatory cytokines. , particularly the p65/p50 heterodimer, binds to the promoter region at positions -105 to -91 in humans and is essential for CCL20 induction by cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1α or IL-1β). Similarly, is activated by interleukin-6 (IL-6) in combination with its soluble receptor (sIL-6R), binding to the CCL20 promoter between -280 and -33 to enhance transcription, particularly in . Activator protein-1 (AP-1), composed of c-Jun/c-Fos, interacts with the promoter at -124 to +33 and contributes to CCL20 upregulation, often in synergy with pathways triggered by TNF-α and IL-1. Epigenetic modifications, including of the CCL20 promoter, serve as a to suppress . Hypermethylation at specific CpG sites inhibits CCL20 transcription by preventing access, while hypomethylation in certain cancers, such as esophageal , leads to aberrant upregulation, highlighting the suppressive role of methylation in normal regulation. Post-transcriptional control of CCL20 involves microRNAs (miRNAs) that target its mRNA for degradation or translational repression. For instance, miR-21 binds directly to the 3' (UTR) of CCL20 mRNA, reducing its stability and expression levels, as demonstrated in cell lines where miR-21 downregulated CCL20 protein production. Environmental factors also modulate CCL20 expression through signaling cascades. Hypoxia induces CCL20 transcription in human monocytes and macrophages via hypoxia-inducible factor-1 (HIF-1) stabilization, promoting chemokine secretion under low-oxygen conditions. Microbial products like lipopolysaccharide (LPS) trigger CCL20 production via Toll-like receptor (TLR) signaling, particularly TLR4 on hepatic stellate cells and macrophages, leading to rapid mRNA upregulation and contributing to inflammatory responses.

Receptor Interaction and Signaling

Binding to CCR6

CCL20, also known as macrophage inflammatory protein-3α (MIP-3α), specifically binds to the G-protein-coupled receptor CCR6, which is predominantly expressed on immature dendritic cells, effector and memory T cells, and B lymphocytes. This receptor-ligand interaction is crucial for directing the migration of these immune cells to sites of inflammation or mucosal tissues. CCL20 serves as the sole high-affinity chemokine ligand for CCR6, with no reported binding to other chemokine receptors, ensuring specificity in its signaling role. The binding affinity between CCL20 and CCR6 is in the nanomolar range, with a dissociation constant (K_D) of approximately 0.9 nM, as determined through radioligand binding assays on transfected cells. This tight interaction is mediated primarily through the N-terminal region and extracellular loops (ECLs), particularly ECL2 and ECL3, of CCR6, where the core domain and N-terminus of CCL20 engage the receptor's orthosteric pocket. Structural studies have provided detailed insights into this binding interface. A cryo-electron microscopy (cryo-EM) structure resolved at 3.3 resolution reveals that CCL20 adopts a compact conformation upon binding, with its globular core interacting with the N-terminus of CCR6 and its N-terminus adopting an extended structure that forms polar interactions, such as salt bridges and hydrogen bonds, within the shallow extracellular binding pocket to stabilize the active receptor state. More recent structural analyses have also elucidated how allosteric antagonists bind to CCR6, inducing conformational changes that prevent CCL20-induced activation. These interactions highlight the molecular basis for the high specificity and potency of CCL20 in activating CCR6.

Downstream Signaling Pathways

Upon binding of CCL20 to its receptor CCR6, a seven-transmembrane (GPCR), the of the Gi/o family is activated, leading to dissociation of the Gαi/o subunit from the Gβγ complex. The Gαi/o subunit inhibits adenylate cyclase, resulting in decreased intracellular cyclic AMP (cAMP) levels. Concurrently, the free Gβγ subunits activate phospholipase C-β (PLC-β), which hydrolyzes (PIP2) into 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), thereby mobilizing intracellular calcium stores and increasing cytosolic Ca²⁺ concentrations. This calcium flux is a critical early event in CCR6-mediated . The activated signaling cascades downstream of CCL20-CCR6 engagement include several key pathways. The PI3K/Akt pathway is stimulated via Gβγ or β-arrestin scaffolds, promoting cell survival and proliferation signals. The MAPK/ERK pathway is rapidly activated, with of ERK1/2 occurring within minutes of binding, facilitating transcriptional responses related to and growth. Additionally, the JAK/ pathway is engaged, particularly through of , which supports production and immune cell differentiation processes. Effector responses involve cytoskeletal reorganization, where the signaling promotes polymerization essential for directed cellular movement. This is mediated in part by Rac , small Rho family GTP-binding proteins activated downstream of PI3K and MAPK, which drive lamellipodia formation and polarity. Signal termination and desensitization occur through receptor by G protein-coupled receptor kinases (GRKs) on specific serine/ residues in the CCR6 C-terminal tail. This recruits β-arrestin proteins, which uncouple the receptor from G proteins, inhibit further signaling, and promote clathrin-mediated internalization of the CCL20-CCR6 complex, thereby attenuating the response.

Biological Functions

Chemotaxis and Cell Recruitment

CCL20 primarily functions as a chemoattractant for specific immune cell subsets through its interaction with the chemokine receptor CCR6, guiding their directional migration via concentration gradients formed in tissues. dendritic cells (DCs), which express high levels of CCR6, are strongly recruited by CCL20 to sites of inflammation or antigen exposure, facilitating their maturation and . Memory T cells, particularly Th17 subsets, also migrate efficiently toward CCL20 gradients, contributing to targeted immune responses at mucosal and inflamed sites. Similarly, IgA-secreting B cells are attracted via the CCL20-CCR6 axis, supporting mucosal immunity in lymphoid tissues. In contrast, CCL20 exhibits only weak chemotactic activity toward neutrophils, despite their presence in inflammatory environments. The mechanism of CCL20-mediated involves sensing by CCR6 on target cells, which triggers intracellular signaling leading to polymerization, polarization, and directed . This process enables cells to navigate complex environments toward higher CCL20 concentrations, often at epithelial or endothelial barriers. studies using transwell migration assays have demonstrated potent recruitment of immature DCs, with half-maximal effective concentrations () typically in the range of 1-30 ng/mL, underscoring CCL20's high-affinity interaction with CCR6. CCL20 gradients are stabilized on the by binding to heparan sulfate proteoglycans (HSPGs), which anchor the to the and cell surfaces, preventing and maintaining localized haptotactic cues for efficient cell recruitment. This immobilization enhances the presentation of CCL20 to circulating CCR6-expressing cells, promoting their and transmigration. Such interactions are critical for directing immune cell infiltration without requiring continuous secretion.

Role in Immune Responses

CCL20 plays a pivotal role in mucosal immunity by facilitating the recruitment of CCR6-expressing T helper 17 (Th17) cells to barrier sites such as the gut and , where they mediate defense against extracellular pathogens. In the intestinal mucosa, CCL20 is constitutively expressed by epithelial cells and M cells in Peyer's patches, promoting the homing of Th17 cells to support steady-state immune surveillance and rapid responses to microbial challenges. This CCL20-CCR6 axis is essential for Th17-mediated production of and cytokines like IL-17 and , which bolster epithelial barrier integrity and recruitment against invaders such as and fungi. Similarly, in the , CCL20 secreted by in response to microbial stimuli or Th17 cytokines attracts CCR6+ Th17 cells, enhancing local defenses at this cutaneous barrier. Beyond mucosal recruitment, CCL20 contributes to adaptive immunity by guiding dendritic cells (DCs) to sites of encounter, thereby supporting their to draining s for T cell priming. Immature DCs expressing CCR6 are drawn by CCL20 gradients produced by epithelial and stromal cells in mucosal tissues, allowing efficient sampling at portals of entry like the gut or . Upon maturation, these DCs upregulate CCR7 for lymph node homing, but initial CCL20-mediated positioning ensures robust to naive T cells, amplifying Th17 and other effector responses. This process is particularly vital in lymphoid structures like Peyer's patches, where CCL20 coordinates DC trafficking to initiate adaptive immunity. In addition to its chemotactic functions, CCL20 exhibits direct activity, contributing to innate host defense by disrupting microbial membranes. This property is effective against such as and , as well as certain parasites and viruses, particularly under low-salt conditions at mucosal surfaces. CCL20 exhibits a dual function in modulating immune balance, attracting both pro-inflammatory Th17 cells and regulatory T cells (Tregs) to fine-tune responses between tolerance and inflammation. While CCL20 drives Th17 infiltration to promote pathogen clearance, it also recruits CCR6+ Foxp3+ Tregs, which produce IL-10 and TGF-β to dampen excessive inflammation and maintain mucosal homeostasis. This bidirectional recruitment helps prevent chronic activation at barrier sites, as seen in the gut where Treg attraction via CCL20 counteracts Th17-driven responses during steady-state conditions. Disruption of this balance can shift toward pathology, but in physiological immunity, it ensures adaptive control. In viral and bacterial infection contexts, CCL20 expression is upregulated to orchestrate targeted immune responses. During infection, epithelial cells in the female reproductive tract and gut produce CCL20 in response to viral exposure, recruiting CCR6+ immune cells while exhibiting direct anti- activity by inhibiting viral entry. For fungal threats like , oral and intestinal epithelial cells rapidly induce CCL20 upon recognition of candidalysin, a fungal , to attract Th17 cells and DCs essential for IL-17-dependent clearance. This upregulation similarly occurs in bacterial challenges, such as , reinforcing mucosal defenses through Th17 polarization.

Role in Diseases

Involvement in Inflammation

CCL20 plays a central role in orchestrating inflammatory responses by attracting immune cells, particularly through its interaction with CCR6 on T helper 17 (Th17) cells, dendritic cells, and neutrophils, thereby amplifying chronic and acute inflammation in various tissues. In autoimmune diseases such as rheumatoid arthritis (RA) and psoriasis, CCL20 expression is markedly elevated, driving pathogenic immune cell infiltration and sustaining tissue damage. For instance, serum and plasma levels of CCL20 are significantly higher in RA patients compared to healthy controls, correlating with disease activity scores and inflammatory markers like C-reactive protein. This chemokine recruits CCR6-expressing mononuclear cells to synovial tissues, enhancing IL-6 production and Th17 cell invasion via the RORγt-CCR6-CCL20 axis, which exacerbates joint inflammation. Similarly, in psoriasis, Th17-derived cytokines such as IL-17A and IL-22 stimulate CCL20 production by epidermal keratinocytes, creating a feedback loop that promotes Th17 cell recruitment to lesional skin and perpetuates psoriatic plaques. Blocking this axis, as demonstrated in preclinical models, reduces psoriasiform dermatitis by limiting Th17 infiltration and cytokine release. In (MS), CCL20 facilitates the recruitment of CCR6+ Th17 cells across the blood-brain barrier, contributing to and demyelination. Elevated serum CCL20 levels are observed in MS patients and correlate with disease activity, with the CCL20-CCR6 axis implicated in Th17-mediated pathogenesis. In airway inflammatory conditions like and (COPD), CCL20 is predominantly produced by airway epithelial cells in response to proinflammatory stimuli, contributing to the CCL20-CCR6 axis that recruits Th17 cells and s to the lungs. Elevated CCL20 levels in asthmatic airways correlate with hypersecretion and Th17-mediated , while in COPD, increased CCL20 expression in bronchial tissues facilitates dendritic cell accumulation and sustains chronic influx. Recent studies highlight the axis's role in severe airway , where epithelial CCL20 upregulation drives immune cell activation and exacerbates airflow obstruction in both conditions. This mechanism underscores CCL20's contribution to persistent lung pathology beyond acute responses. In (IBD), particularly , CCL20 is overexpressed in colonic epithelium and , promoting the homing of CCR6+ and dendritic cells to the gut mucosa, which amplifies mucosal inflammation. Biopsies from active show significantly higher CCL20 mRNA and protein levels compared to inactive disease or controls, correlating with disease severity and Th17 cell infiltration. The CCL20-CCR6 pathway facilitates retention and activation in the inflamed intestine, contributing to barrier dysfunction and ; preclinical blockade of this axis reduces immune cell homing and attenuates experimental . further elevates CCL20 in patients, linking nutritional status to heightened inflammatory homing. In kidney diseases, the CCL20-CCR6 axis promotes tubular epithelial injury and immune recruitment, playing a pivotal role in the progression from (AKI) to (CKD). Elevated circulating and urinary CCL20 levels are associated with in renal tissues, where it attracts Th17 cells and other CCR6+ leukocytes, exacerbating damage as observed in and preclinical models as of 2024. During acute , such as in bacterial , CCL20 mediates recruitment to sites, enhancing early but potentially contributing to excessive tissue damage if dysregulated. In models of and , CCL20-CCR6 signaling is critical for and activation, with CCL20 neutralization increasing mortality by impairing bacterial clearance in the gut and lungs. Epithelial cells at sites rapidly produce CCL20 in response to bacterial agonists, directing CCR6-expressing neutrophils to contain pathogens like or , though this can lead to amplified in vulnerable tissues.

Role in Cancer

CCL20 is expressed by various cancer cells, including those in colorectal and breast tumors, where it facilitates the recruitment of immunosuppressive cells such as CCR6-expressing regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) to the tumor microenvironment, thereby promoting immune evasion and tumor progression. In colorectal cancer, tumor-derived CCL20 specifically attracts Tregs, enhancing chemoresistance through pathways involving FOXO1/CEBPB/NF-κB signaling. Similarly, in breast cancer, CCL20 secreted by cancer cells modulates polymorphonuclear MDSCs (PMN-MDSCs), fostering cancer stemness and metastasis via ERK1/2-Sox2 signaling. The CCL20-CCR6 axis contributes to metastasis by promoting cancer cell homing to distant sites, including lymph nodes and the liver, and is associated with poor clinical outcomes. In non-small cell lung cancer, elevated CCL20 expression correlates with advanced tumor stages, increased lymph node metastasis, and reduced patient survival. For colorectal cancer, high CCL20 levels synergize with CXCL8 to drive liver metastasis and predict unfavorable prognosis. Across multiple cancers, including breast and colorectal, upregulated CCL20 expression is linked to lower overall and metastasis-free survival rates. A 2020 review highlights the CCL20-CCR6 axis's role in driving tumor progression through direct enhancement of and , as well as indirect remodeling of the in cancers such as , breast, and colorectal. This axis also promotes Th17 cell polarization and recruitment to tumor sites, as observed in cervical and colorectal cancers, where CCL20-CCR6 signaling facilitates Th17 infiltration, potentially exacerbating and disease advancement. While CCL20 can recruit immature dendritic cells (DCs) capable of to initiate anti-tumor responses, its predominant effect in the cancer context is pro-tumorigenic, as it preferentially attracts suppressive Tregs and MDSCs that inhibit effector immune functions. This dual role underscores the context-dependent nature of CCL20 in modulating anti-tumor immunity versus immune evasion.

Clinical and Research Implications

Diagnostic and Prognostic Value

CCL20 has emerged as a potential biomarker for diagnosing and prognosticating inflammatory and neoplastic conditions due to its measurable levels in biological fluids and tissues. In serum and plasma, elevated CCL20 concentrations are observed in patients with psoriasis, where levels correlate with disease severity and systemic inflammation, outperforming markers like IL-6 and IL-17A in reflecting vascular endothelial involvement. Similarly, plasma CCL20 is associated with increased risk of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, as identified in large-scale proteomic analyses. Recent Olink proteomics studies, deposited in NCBI databases in 2025, highlight CCL20's utility for early detection of inflammatory disorders by quantifying its upregulation in at-risk populations prior to clinical onset. Tissue expression of CCL20, assessed via (IHC) staining, serves as a prognostic indicator in various tumors, particularly for risk. In , high CCL20 expression in tumor-associated macrophages correlates with aggressive phenotypes, reduced overall , and higher metastatic potential, as evidenced by IHC analyses of primary lesions. Likewise, elevated CCL20 in stroma predicts metastatic progression and poorer patient outcomes, with IHC revealing its localization in immune infiltrates that facilitate tumor spread. Prognostic value is further supported by CCL20 mRNA levels in (CRC), where upregulation is linked to advanced disease stages. High CCL20 mRNA expression is notably increased in stage IV CRC compared to earlier stages, associating with and chemoresistance through recruitment of regulatory T cells. Serum CCL20 levels, often combined with IL-17A, independently predict CRC progression and survival, offering a non-invasive prognostic panel. Despite these associations, CCL20's broad role in limits its specificity as a standalone , as elevated levels occur across multiple conditions like , IBD, and various cancers, potentially confounding disease-specific diagnostics. This overlap necessitates approaches, such as combining CCL20 with other markers, to enhance diagnostic accuracy.

Therapeutic Targeting

Therapeutic strategies targeting the CCL20-CCR6 axis aim to disrupt chemokine-mediated recruitment of immune cells, particularly Th17 cells and dendritic cells, in inflammatory and neoplastic conditions. inhibitors and monoclonal antibodies directed at CCR6 have shown promise in preclinical models by blocking ligand binding and downstream signaling, thereby reducing pathological inflammation without broadly suppressing immunity. For instance, the allosteric CCR6 PF-07054894, a squaramide-based compound, potently inhibits CCL20-induced of T cells and has advanced to Phase I clinical trials for , demonstrating favorable and safety in early studies. Similarly, the oral CCR6 inhibitor IDOR-1117-2520 dose-dependently attenuates infiltration of CCR6+ immune cells in models of skin inflammation, exhibiting efficacy comparable to IL-17 and IL-23 inhibitors, with potential extension to (IBD) and cancer. Anti-CCR6 monoclonal antibodies represent another key class of inhibitors under , particularly for IBD and cancer. Preclinical studies with anti-CCR6 antibodies have shown of CCR6-mediated immune in models of inflammation and tumor progression. In models, CCR6 via antibodies or antagonists suppresses CCL20-driven progression, highlighting the axis's role in remodeling. Neutralizing antibodies against CCL20 have been evaluated primarily in and related autoimmune models. The GSK3050002, a humanized IgG1κ with high for CCL20, effectively blocks receptor activation and has completed Phase I trials in healthy volunteers, showing good tolerability and dose-dependent target engagement. In preclinical models, anti-CCL20 antibodies reduce epidermal trafficking of γδ T cells and production, alleviating psoriasiform in IL-23-induced mice. The planned Phase II trial of GSK3050002 in (NCT02671188) was withdrawn before enrollment, with development paused due to immune complex formation observed in preclinical monkey studies. Gene therapy approaches, including siRNA-mediated knockdown, offer targeted modulation of CCL20 expression in inflammatory settings. In models of airway , siRNA targeting upstream regulators like S1PR3 suppresses CCL20 production in bronchial epithelial cells, attenuating Th17 and responses. A 2024 review in the Journal of highlights siRNA strategies as emerging tools for silencing CCL20 in respiratory diseases, with preclinical data showing reduced airway hyperresponsiveness in models following CCL20 knockdown. These nucleic acid-based therapies provide spatiotemporal control but require delivery optimizations for clinical translation. Despite these advances, therapeutic targeting of CCL20-CCR6 faces challenges, including off-target effects on mucosal immunity, where the axis is essential for homeostatic recruitment and barrier defense. Inhibition may impair responses to pathogens at mucosal sites, as evidenced by increased in CCR6-deficient models. Additionally, immune formation with anti-CCL20 antibodies poses toxicity risks, as seen in preclinical studies. As of 2025, ongoing I/II trials, such as those for PF-07054894 in IBD and exploratory anti-CCR6 agents in solid tumors, underscore the need for refined selectivity to balance efficacy and safety.