Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Sep 12.
Published in final edited form as: Postdoc J. 2014 Mar;2(3):7–16.

Many Roles of CCL20: Emphasis on Breast Cancer

Kingsley O Osuala 1,*, Bonnie F Sloane 1
PMCID: PMC5019369  NIHMSID: NIHMS810788  PMID: 27631019

Abstract

CCL20 or MIP3α is a small ~8 kDa protein primarily expressed in the liver, colon, prostate, cervix, and skin. The cellular receptor for CCL20 is CCR6. CCl20 unlike many other cytokines only binds CCR6, making the CCL20/CCR6 pathway an attractive drug target. Since the initial discovery of CCL20 in the early 1990's, there has been an increase in the evidence implicating the chemokine and its receptor in a number of diseases, including rheumatoid arthritis and human immunodeficiency virus infection. CCL20 has also been linked to malignancies such as ovarian, colorectal and pancreatic cancers. CCL20 can also attract tumor-promoting immune-suppressive cells to the tumor microenvironment, which may contribute to the immune evasive potential of the tumor and tumor progression.

Keywords: breast cancer, CCL20, CCR6, chemokines, MIP3α

Introduction

Advances in our basic scientific understanding of cancer have led to a clear link between the progression of cancer and the infiltration of immune cells. Understanding how immune response propagates tumor development has become a field of study all its own. Recent studies of the tumor microenvironment (TME) have expanded our knowledge about the involvement of the immune system in tumor progression [1, 2]. Many of these studies have shown a correlation between the infiltration of immune cells (primarily CD8+ and CD4+ T-lymphocytes and macrophages,), tumor burden and patient survival [3, 4].

In a number of cancers, cytokines are significantly upregulated [3, 5], adding to a complex communication network within the TME and systemic recruitment of immune responding cells. It seems apparent that tumor cells utilize autocrine/paracrine-signaling mechanisms to promote cell survival by inducing a shift in gene and protein expression among themselves and neighboring cells. This in turn, facilitates positive feedback loops, which enhance production and secretion of many pro-survival cytokines, proteases, and growth factors [6, 7].

Chemokines and their receptors are exciting targets for therapeutics with several drugs targeting chemokine pathways having reached clinical trials [8-10]. To date, more than 51 chemokines have been identified, and only a subset of these have been studied in depth. Chemokines in general are divided into 4 major groups based on N-terminal arrangement of conserved cysteine residues. The CC chemokines represent the largest of the four groups with 28 identified members. Receptors of the CC family of chemokines are G-protein coupled receptors expressed in a variety of inflammatory trafficking cells and some cancer cells [11]. In many cases, chemokine secreting cells also express the concomitant chemokine receptors and hence can function in a paracrine and/or autocrine manner [12-15].

CCL20

CCL20, also known as macrophage inflammatory protein 3 alpha (MIP3α) [16], Liver and Activation Regulated Chemokine (LARC) [17], , and Exodus-1 [18], was first discovered and characterized in hepatocytes [17] and later shown to be expressed in the lung [19] and various connective and lymphatic tissues [20, 21]. The gene encoding CCL20 was mapped to chromosome 2q and contains 4 exons and 3 introns [17, 22]. This is a slight variation from most CC chemokine gene sequences that contain 3 exons and 2 introns. The gene contains many transcription factor-binding sites including NFkB, AP-1 and 2, C-EBP, ETS, and SP1 [23-27].

The full-length pro-CCL20 is 96 amino acids in length and contains a classical N-terminal signal sequence, which is cleaved to yield the mature peptide sequence of 70 amino acids (CCL20, 1-70). A single in-frame deletion of alanine at position 27 in the pro-peptide results in a fully functional variant of the full-length protein (CCL20, 2-70) [19, 22]. The mature translated protein contains 2 cysteine-based disulfide bridges that form a “Greek Key” motif in its tertiary structure (Fig.1).

Figure 1.

Figure 1

“Greek Key” motif. (A) A representative illustration of the “meandros” art pattern, commonly known as the Greek key (B) The antiparallel strands of the CCL20 protein loop to form a structure reminiscent of the artistic Greek key. The strands are bound together by cysteine disulfide bonds, and CC chemokines have a classical pair of cysteine residues near their amino terminus (red arrow).

Hoover et al. resolved the X-ray crystal structure of human CCL20 [28], and Perez-Canadillas and Chan defined the nuclear magnetic resonance structure [29, 30]. These studies revealed the underlying structural scheme of CCL20, which confers the chemokine innate antimicrobial activity. The groups showed that CCL20 contains a carboxy-terminal alpha helix loop structure consisting of several positively charged residues and yielding a highly cationic region of the peptide. Such cationic properties are commonly associated with naturally occurring antimicrobial peptides such as human lactoferrin [31, 32] and the defensins [28, 33, 34]. CCL20 likely plays an important role in innate immune defense as its expression is induced at common sites of infection, including gastric mucosa and respiratory epithelium [35, 36].

Chan et al. showed that CCL20 existed as a monomeric protein in solutions of pH 3.5 and 4.6, and as a homodimer in solutions of pH 7.0 and 7.5. These data suggest that pH may regulate the structure and bioactivity of CCL20 [30]. This would allow for alternate modes of action for CCL20 based on local pH although no data are yet available to support such functions [37, 38].

CCL20 Expression

As a chemoattractant, CCL20 plays a crucial role in the recruitment of CD34+ derived dendritic cells and T cells [36, 39-41]. Accordingly, upregulation of CCL20 mRNA and protein upon pro-inflammatory stimuli has been shown in various mouse and human tissues [27, 42, 43]. Many pro-inflammatory chemokines and small molecules mediate downstream signaling through activation of the NFkB pathway. This is also true for CCL20, as studies have shown that its expression can be altered by effectors of NFkB signaling namely: TNFα, IL-1α and beta, IL6, and IL-17 [24, 42, 44-46].

CCL20 Receptor

The receptor for CCL20 is a 7-transmembrane G-protein coupled receptor of the beta chemokine family [47]. Initially acknowledged as an orphan receptor and given several different names including: GPR-CY4, STRL22 [48], and CKRL-3 [49]. The receptor was designated CCR6 in 1997 by Baba et al. [21]. CCR6 is one of 11 identified receptors belonging to the CC family of chemokine receptors. The only ligands known to bind to CCR6 are β-defensins 1 and 2 and CCL20 [50]. CCR6 shows low-level mRNA and protein expression in most tissues under non-pathological conditions; intestinal mucosa, lung mucosa and lymphoid tissues have the highest levels of expression [51-53]. At the cellular level CCR6 is primarily expressed in Th17 [54], T-reg cells [47], immature dendritic cells [19, 39, 55], subsets of CD8+ cytotoxic T cells [56], memory and effector T cells , and B cells [57]. Basal expression of CCR6 on immune cells is utilized for cellular homing to sites of ligand secretion [47, 56, 58]. In pathological autoimmune diseases, such as inflammatory bowel disease and rheumatoid arthritis, CCR6 is significantly upregulated [59-61]. Interestingly, some epithelial tumors have also been found to express CCR6 [62].

CCL20: Rheumatoid Arthritis and HIV

CCL20 is a key player in the recruitment of inflammatory cells, implicating the chemokine in a variety of inflammatory diseases. Matsui et al. showed that synovial fluid from rheumatoid arthritis (RA) patients had increased concentrations of CCL20 compared to synovial fluid from patients with osteoarthritis [63]. To date multiple studies have established a link between CCL20 and the infiltration of CCR6+ dendritic cells, macrophages and CD4+ T cells to synovial joints [43, 45]. Murakami et al. have shown that recruitment of CD4+ cells to synovial joints in a mouse model of rheumatoid arthritis is a key step in the development of this autoimmune disease [64]. The mechanism of recruitment was identified as an IL-6 and IL-17 mediated expression of chemokines, in particular of CCL20.

The CCL20/CCR6 axis has also been associated with human immunodeficiency virus (HIV) infection. A report by Gosselin et al. suggests that CCR6 is involved in the HIV infection process as CCR6+ T cells showed a significantly higher rate of HIV infectivity vs. CCR6- T cells [65]. Another group has shown that CCL20, CCL19, and CXCL10 can induce latent infection of resting activated CD4+ T cells. This provided a mechanism for a marked reinfection, as these chemokines can recruit CD4+ cells to the blood stream where they target HIV virus laden lymphoid tissues [66].

In contrast to the cooperative function of CCR6 in HIV infection, CCL20 acts as an antiviral agent against HIV [67]. Ghosh et.al discovered that CCL20 directly inhibited HIV infection of TZM-bl cells when recombinant CCL20 was preincubated with the virus before first round infection of cell cultures [67]. A study by Fontaine et al. revealed elevated steady state levels of CCL20, CCL2, and CCL19 in blood samples from HIV+ patients [68]. These data support a role for CCL20 in innate immunity. Additionally, many studies have shown that CCL20 is secreted by neutrophils as a first response to infection and that increased CCL20 recruits dendritic cells, thereby implicating CCL20 in the initiation of acquired immunity [69, 70].

CCL20 and Breast Cancer

In recent years, the immune response associated with developing tumors has become a hot topic of study. Chemokines being primary effectors of inflammation correspondingly play a role in immune response. Chemokines have been linked to several types of malignancies, including prostate [71], colorectal [72, 73], ovarian [74, 75], and breast cancer [3, 76]. This is an expected linkage since many tumors generate an inflammatory response [77, 78].

Dendritic cells

CCL20 and its receptor CCR6 may have a significant functional role in the progression and invasion of breast carcinomas. Although there is a limited understanding of the specific contribution of CCL20 in breast cancer development, CCL20 can recruit CCR6-expressing dendritic cells into epithelial tissues [40] and the infiltration of dendritic cells into tumors or surrounding stroma has been associated with poor prognosis [79]. Bonnotte et al. showed that infiltrating immune cells of colon adenocarcinoma in female rats did not develop an antitumor phenotype as these cells failed to acquire the ability to activate T cells [80]. The mechanism by which dendritic cells retain an immature phenotype in a tumor antigen-rich environment is unknown; however, upregulation of tumor promoting genes may play a role [81]. This phenomenon has also been observed systemically in cancer patients [82], suggesting a significant tumor-mediated reprogramming of dendritic cell function. Le Mercier et al., showed that the pro-tumorigenic/immature phenotype of tumor-associated dendritic cells might be a result of the loss of Toll-like receptor 7 (TLR7) activation. Intra-tumoral injection of TLR7 ligand in an orthotopic mammary tumor model has been shown to be sufficient to induce a shift in the cytokine signature of the tumors and their regression [83]. These data provide a possible mechanism by which dendritic cells are reprogrammed to support tumor immune evasion and tumor progression.

Recently, Marsigliante et al. reported that CCL20 induces migration of human breast cancer cells through activation of both Akt and mitogen-activated protein kinase pathways [84]. Both pathways have been shown to promote tumor growth and metastasis [85, 86]. In addition, CCL20 induces the expression of MMP-9 via NFκB activation [84]. These data are consistent with CCL20 playing a role in both tumor cell survival and metastasis.

Tumor-Associated Macrophages

Tumor-associated macrophages (TAMs) are directly involved in cancer progression [87]. TAMs are pro-neoplastic and have a pro-inflammatory gene signature [88], which result in an upregulation of cytokines and the recruitment of immune cells to the TME (Fig. 2). To date a substantial body of evidence supports the involvement of TAMs in the progression and metastasis of breast cancer [89-91]. Bierie et al., showed data that altered TGFβ signaling in the breast tumor microenvironment can upregulate CCL20 in breast tumor cells and that secretion of proinflammatory cytokines may promote early tumor progression [92].

Figure 2.

Figure 2

Chemoattractant capability of CCL20. (A) Breast tumor cells expressing CCL20 can attract CCR6 expressing immune cells to the tumor microenvironment, therefore facilitating breakdown of the extracellular matrix. (B) Breast tumor cells that break through the extracellular matrix can migrate to and invade nearby blood vessels. Once in the blood stream tumor cells expressing CCR6 can home to lymph nodes or other organ sites that secrete CCL20.

TAMs overexpress and secrete proteases, such as those of the matrix metalloproteinase family, cysteine cathepsins, and serine proteases [90, 93, 94], into the TME. The cysteine cathepsin B and the aspartic cathepsin D have been shown to differentially cleave the monomeric peptide of CCL20 in vitro yielding either a fully functional CCL20 or a chemoattractant null peptide, respectively [95]. Both of these lysosomal proteases are frequently upregulated in pathological conditions including breast cancer [96-99]. The altered expression of cathepsin B occurs at the message and protein level and results in an increase in cell surface and soluble secreted cathepsin B [100]. As CCL20 is a secreted chemokine there may be an extracellular interface of protease and chemokine. The distinct in vivo relationship between cathepsin B and CCL20 has yet to be studied.

Micro RNAs

The study of micro RNAs (miRNAs) has redefined our understanding of gene regulation and revealed novel regulatory functions for small non-translated RNAs [101]. The introduction of next generation sequencing modalities has rapidly identified several miRNAs, including miR-205, miR-125b, miR-145, and miR-21, which are differentially expressed in breast carcinomas vs. normal breast tissue [102-104]. Interestingly, miR-21 binds to the 3’ untranslated region of CCL20 mRNA initiating its degradation [105]. Terao et al. found miR-21 to be differentially expressed in the MCF-7 human breast cancer cell line, and to have different downstream effects in estrogen receptor positive versus estrogen receptor negative breast cancer cell lines [106].

In summation a specific role for CCL20 in breast cancer has not yet been clearly established. Many studies discussed here indicate that CCL20 is deregulated in breast cancer, but to what extent and in what capacity CCL20 may contribute to tumorigenicity is not known [107, 108]. Moving forward, we should evaluate the functional role of CCL20 and how signaling through CCR6 affects gene transcription. These studies should be followed with assays that can monitor functional changes in cell behavior as a result of CCL20 mediated CCR6 activation. Further studies should examine the effects of up-regulation of CCL20 in an in vivo context where an immune cell response can be monitored. This would require the use of animal models with an intact immune system or a humanized immune system. Additionally, examining the effects of CCL20 on immune cell receptor presentation will aid in our understanding of how CCL20 might alter immune cell function. Results from such studies will be invaluable for understanding the role of CCL20 in breast cancer and given the selectivity of CCL20 for its receptor CCR6, therapeutics targeting the ligand/receptor interaction will be relatively easy to test.

Acknowledgments

This work was supported in part by the Department of Defense Breast Cancer Research Program Postdoctoral Fellowship Award (W81XWH-12-1-0024) and R01 CA131990 from the National Institutes of Health.

References

  • 1.María Apellániz-Ruiz ZC. Adapt or Dye: Tumor Microenvironment, A Powerful Regulator of Cancer Progression. PostDoc Journal. 2013;1(5) [Google Scholar]
  • 2.Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309–22. doi: 10.1016/j.ccr.2012.02.022. [DOI] [PubMed] [Google Scholar]
  • 3.Bell D, et al. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med. 1999;190(10):1417–26. doi: 10.1084/jem.190.10.1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cassier PA, et al. Prognostic value of the expression of C-Chemokine Receptor 6 and 7 and their ligands in non-metastatic breast cancer. BMC Cancer. 2011;11:213. doi: 10.1186/1471-2407-11-213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Dellacasagrande J, et al. Liver metastasis of cancer facilitated by chemokine receptor CCR6. Scand J Immunol. 2003;57(6):534–44. doi: 10.1046/j.1365-3083.2003.01263.x. [DOI] [PubMed] [Google Scholar]
  • 6.Kim KY, et al. Adipocyte culture medium stimulates invasiveness of MDA-MB-231 cell via CCL20 production. Oncol Rep. 2009;22(6):1497–504. doi: 10.3892/or_00000593. [DOI] [PubMed] [Google Scholar]
  • 7.Marrogi AJ, et al. Study of tumor infiltrating lymphocytes and transforming growth factor-beta as prognostic factors in breast carcinoma. Int J Cancer. 1997;74(5):492–501. doi: 10.1002/(sici)1097-0215(19971021)74:5<492::aid-ijc3>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 8.Reckless J, Grainger DJ. Identification of oligopeptide sequences which inhibit migration induced by a wide range of chemokines. Biochem J. 1999;340(Pt 3):803–11. [PMC free article] [PubMed] [Google Scholar]
  • 9.Grainger DJ, Reckless J, Fox DJ. Broad spectrum chemokine inhibitors related to NR58-3.14.3. Mini Rev Med Chem. 2005;5(9):825–32. doi: 10.2174/1389557054867101. [DOI] [PubMed] [Google Scholar]
  • 10.Miklos S, et al. Preventive usage of broad spectrum chemokine inhibitor NR58-3.14.3 reduces the severity of pulmonary and hepatic graft-versus-host disease. Int J Hematol. 2009;89(3):383–97. doi: 10.1007/s12185-009-0272-y. [DOI] [PubMed] [Google Scholar]
  • 11.Balkwill FR. The chemokine system and cancer. J Pathol. 2012;226(2):148–57. doi: 10.1002/path.3029. [DOI] [PubMed] [Google Scholar]
  • 12.Nicolson GL. Cancer progression and growth: relationship of paracrine and autocrine growth mechanisms to organ preference of metastasis. Exp Cell Res. 1993;204(2):171–80. doi: 10.1006/excr.1993.1022. [DOI] [PubMed] [Google Scholar]
  • 13.Fang WB, et al. CCL2/CCR2 chemokine signaling coordinates survival and motility of breast cancer cells through Smad3 protein- and p42/44 mitogen-activated protein kinase (MAPK)-dependent mechanisms. J Biol Chem. 2012;287(43):36593–608. doi: 10.1074/jbc.M112.365999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hsu CJ, et al. AMP-activated protein kinase activation mediates CCL3-induced cell migration and matrix metalloproteinase-2 expression in human chondrosarcoma. Cell Commun Signal. 2013;11:68. doi: 10.1186/1478-811X-11-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Frick VO, et al. CCR6/CCL20 chemokine expression profile in distinct colorectal malignancies. Scand J Immunol. 2013;78(3):298–305. doi: 10.1111/sji.12087. [DOI] [PubMed] [Google Scholar]
  • 16.Rossi DL, et al. Identification through bioinformatics of two new macrophage proinflammatory human chemokines: MIP-3alpha and MIP-3beta. J Immunol. 1997;158(3):1033–6. [PubMed] [Google Scholar]
  • 17.Hieshima K, et al. Molecular cloning of a novel human CC chemokine liver and activation-regulated chemokine (LARC) expressed in liver. Chemotactic activity for lymphocytes and gene localization on chromosome 2. J Biol Chem. 1997;272(9):5846–53. doi: 10.1074/jbc.272.9.5846. [DOI] [PubMed] [Google Scholar]
  • 18.Hromas R, et al. Cloning and characterization of exodus, a novel beta-chemokine. Blood. 1997;89(9):3315–22. [PubMed] [Google Scholar]
  • 19.Power CA, et al. Cloning and characterization of a specific receptor for the novel CC chemokine MIP-3alpha from lung dendritic cells. J Exp Med. 1997;186(6):825–35. doi: 10.1084/jem.186.6.825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cremel M, et al. Characterization of CCL20 secretion by human epithelial vaginal cells: involvement in Langerhans cell precursor attraction. J Leukoc Biol. 2005;78(1):158–66. doi: 10.1189/jlb.0305147. [DOI] [PubMed] [Google Scholar]
  • 21.Baba M, et al. Identification of CCR6, the specific receptor for a novel lymphocyte-directed CC chemokine LARC. J Biol Chem. 1997;272(23):14893–8. doi: 10.1074/jbc.272.23.14893. [DOI] [PubMed] [Google Scholar]
  • 22.Nelson RT, et al. Genomic organization of the CC chemokine mip-3alpha/CCL20/larc/exodus/SCYA20, showing gene structure, splice variants, and chromosome localization. Genomics. 2001;73(1):28–37. doi: 10.1006/geno.2001.6482. [DOI] [PubMed] [Google Scholar]
  • 23.Kwon JH, et al. ESE-1, an enterocyte-specific Ets transcription factor, regulates MIP-3alpha gene expression in Caco-2 human colonic epithelial cells. J Biol Chem. 2003;278(2):875–84. doi: 10.1074/jbc.M208241200. [DOI] [PubMed] [Google Scholar]
  • 24.Harant H, Eldershaw SA, Lindley IJ. Human macrophage inflammatory protein-3alpha/CCL20/LARC/Exodus/SCYA20 is transcriptionally upregulated by tumor necrosis factor-alpha via a non-standard NF-kappaB site. FEBS Lett. 2001;509(3):439–45. doi: 10.1016/s0014-5793(01)03138-6. [DOI] [PubMed] [Google Scholar]
  • 25.Rhee SH, et al. MEK is a key modulator for TLR5-induced interleukin-8 and MIP3alpha gene expression in non-transformed human colonic epithelial cells. J Biol Chem. 2004;279(24):25179–88. doi: 10.1074/jbc.M400967200. [DOI] [PubMed] [Google Scholar]
  • 26.Varesio L, et al. Macrophage-inflammatory protein-3alpha/CCL-20 is transcriptionally induced by the iron chelator desferrioxamine in human mononuclear phagocytes through nuclear factor (NF)-kappaB. Mol Immunol. 2010;47(4):685–93. doi: 10.1016/j.molimm.2009.10.031. [DOI] [PubMed] [Google Scholar]
  • 27.Sugita S, et al. Induction of macrophage-inflammatory protein-3alpha gene expression by TNF-dependent NF-kappaB activation. J Immunol. 2002;168(11):5621–8. doi: 10.4049/jimmunol.168.11.5621. [DOI] [PubMed] [Google Scholar]
  • 28.Hoover DM, et al. The structure of human macrophage inflammatory protein-3alpha /CCL20. Linking antimicrobial and CC chemokine receptor-6-binding activities with human beta-defensins. J Biol Chem. 2002;277(40):37647–54. doi: 10.1074/jbc.M203907200. [DOI] [PubMed] [Google Scholar]
  • 29.Perez-Canadillas JM, et al. NMR solution structure of murine CCL20/MIP-3alpha, a chemokine that specifically chemoattracts immature dendritic cells and lymphocytes through its highly specific interaction with the beta-chemokine receptor CCR6. J Biol Chem. 2001;276(30):28372–9. doi: 10.1074/jbc.M103121200. [DOI] [PubMed] [Google Scholar]
  • 30.Chan DI, et al. Human macrophage inflammatory protein 3alpha: protein and peptide nuclear magnetic resonance solution structures, dimerization, dynamics, and anti-infective properties. Antimicrob Agents Chemother. 2008;52(3):883–94. doi: 10.1128/AAC.00805-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Arnold RR, et al. Bactericidal activity of human lactoferrin: differentiation from the stasis of iron deprivation. Infect Immun. 1982;35(3):792–9. doi: 10.1128/iai.35.3.792-799.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gonzalez-Chavez SA, Arevalo-Gallegos S, Rascon-Cruz Q. Lactoferrin: structure, function and applications. Int J Antimicrob Agents. 2009;33(4):301 e1–8. doi: 10.1016/j.ijantimicag.2008.07.020. [DOI] [PubMed] [Google Scholar]
  • 33.Lehrer RI, Lichtenstein AK, Ganz T. Defensins: antimicrobial and cytotoxic peptides of mammalian cells. Annu Rev Immunol. 1993;11:105–28. doi: 10.1146/annurev.iy.11.040193.000541. [DOI] [PubMed] [Google Scholar]
  • 34.Ganz T, Selsted ME, Lehrer RI. Defensins. Eur J Haematol. 1990;44(1):1–8. doi: 10.1111/j.1600-0609.1990.tb00339.x. [DOI] [PubMed] [Google Scholar]
  • 35.Sierro F, et al. Flagellin stimulation of intestinal epithelial cells triggers CCL20-mediated migration of dendritic cells. Proc Natl Acad Sci U S A. 2001;98(24):13722–7. doi: 10.1073/pnas.241308598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Reibman J, et al. Airway epithelial cells release MIP-3alpha/CCL20 in response to cytokines and ambient particulate matter. Am J Respir Cell Mol Biol. 2003;28(6):648–54. doi: 10.1165/rcmb.2002-0095OC. [DOI] [PubMed] [Google Scholar]
  • 37.Gerweck LE, Seetharaman K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 1996;56(6):1194–8. [PubMed] [Google Scholar]
  • 38.Lutz NW, et al. Quantitative in vivo characterization of intracellular and extracellular pH profiles in heterogeneous tumors: a novel method enabling multiparametric pH analysis. Cancer Res. 2013;73(15):4616–28. doi: 10.1158/0008-5472.CAN-13-0767. [DOI] [PubMed] [Google Scholar]
  • 39.Greaves DR, et al. CCR6, a CC chemokine receptor that interacts with macrophage inflammatory protein 3alpha and is highly expressed in human dendritic cells. J Exp Med. 1997;186(6):837–44. doi: 10.1084/jem.186.6.837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Le Borgne M, et al. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity. 2006;24(2):191–201. doi: 10.1016/j.immuni.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 41.Cook KW, et al. CCL20/CCR6-mediated migration of regulatory T cells to the Helicobacter pylori-infected human gastric mucosa. Gut. 2014 doi: 10.1136/gutjnl-2013-306253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kao CY, et al. Up-regulation of CC chemokine ligand 20 expression in human airway epithelium by IL-17 through a JAK-independent but MEK/NF-kappaB-dependent signaling pathway. J Immunol. 2005;175(10):6676–85. doi: 10.4049/jimmunol.175.10.6676. [DOI] [PubMed] [Google Scholar]
  • 43.Chabaud M, Page G, Miossec P. Enhancing effect of IL-1, IL-17, and TNF-alpha on macrophage inflammatory protein-3alpha production in rheumatoid arthritis: regulation by soluble receptors and Th2 cytokines. J Immunol. 2001;167(10):6015–20. doi: 10.4049/jimmunol.167.10.6015. [DOI] [PubMed] [Google Scholar]
  • 44.Erez N, et al. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell. 2010;17(2):135–47. doi: 10.1016/j.ccr.2009.12.041. [DOI] [PubMed] [Google Scholar]
  • 45.Chevrel G, Garnero P, Miossec P. Addition of interleukin 1 (IL1) and IL17 soluble receptors to a tumour necrosis factor alpha soluble receptor more effectively reduces the production of IL6 and macrophage inhibitory protein-3alpha and increases that of collagen in an in vitro model of rheumatoid synoviocyte activation. Ann Rheum Dis. 2002;61(8):730–3. doi: 10.1136/ard.61.8.730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Berlier W, et al. Seminal plasma promotes the attraction of Langerhans cells via the secretion of CCL20 by vaginal epithelial cells: involvement in the sexual transmission of HIV. Hum Reprod. 2006;21(5):1135–42. doi: 10.1093/humrep/dei496. [DOI] [PubMed] [Google Scholar]
  • 47.Yamazaki T, et al. CCR6 regulates the migration of inflammatory and regulatory T cells. J Immunol. 2008;181(12):8391–401. doi: 10.4049/jimmunol.181.12.8391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Liao F, et al. STRL22 is a receptor for the CC chemokine MIP-3alpha. Biochem Biophys Res Commun. 1997;236(1):212–7. doi: 10.1006/bbrc.1997.6936. [DOI] [PubMed] [Google Scholar]
  • 49.Zaballos A, et al. Molecular cloning and RNA expression of two new human chemokine receptor-like genes. Biochem Biophys Res Commun. 1996;227(3):846–53. doi: 10.1006/bbrc.1996.1595. [DOI] [PubMed] [Google Scholar]
  • 50.Yang D, et al. Beta-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science. 1999;286(5439):525–8. doi: 10.1126/science.286.5439.525. [DOI] [PubMed] [Google Scholar]
  • 51.Cook DN, et al. CCR6 mediates dendritic cell localization, lymphocyte homeostasis, and immune responses in mucosal tissue. Immunity. 2000;12(5):495–503. doi: 10.1016/s1074-7613(00)80201-0. [DOI] [PubMed] [Google Scholar]
  • 52.Williams IR. CCR6 and CCL20: partners in intestinal immunity and lymphorganogenesis. Ann N Y Acad Sci. 2006;1072:52–61. doi: 10.1196/annals.1326.036. [DOI] [PubMed] [Google Scholar]
  • 53.Acosta-Rodriguez EV, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. 2007;8(6):639–46. doi: 10.1038/ni1467. [DOI] [PubMed] [Google Scholar]
  • 54.Annunziato F, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. 2007;204(8):1849–61. doi: 10.1084/jem.20070663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Charbonnier AS, et al. Macrophage inflammatory protein 3alpha is involved in the constitutive trafficking of epidermal langerhans cells. J Exp Med. 1999;190(12):1755–68. doi: 10.1084/jem.190.12.1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kondo T, Takata H, Takiguchi M. Functional expression of chemokine receptor CCR6 on human effector memory CD8+ T cells. Eur J Immunol. 2007;37(1):54–65. doi: 10.1002/eji.200636251. [DOI] [PubMed] [Google Scholar]
  • 57.Krzysiek R, et al. Regulation of CCR6 chemokine receptor expression and responsiveness to macrophage inflammatory protein-3alpha/CCL20 in human B cells. Blood. 2000;96(7):2338–45. [PubMed] [Google Scholar]
  • 58.Varona R, et al. CCR6-deficient mice have impaired leukocyte homeostasis and altered contact hypersensitivity and delayed-type hypersensitivity responses. J Clin Invest. 2001;107(6):R37–45. doi: 10.1172/JCI11297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Varona R, et al. CCR6 has a non-redundant role in the development of inflammatory bowel disease. Eur J Immunol. 2003;33(10):2937–46. doi: 10.1002/eji.200324347. [DOI] [PubMed] [Google Scholar]
  • 60.Kaser A, et al. Increased expression of CCL20 in human inflammatory bowel disease. J Clin Immunol. 2004;24(1):74–85. doi: 10.1023/B:JOCI.0000018066.46279.6b. [DOI] [PubMed] [Google Scholar]
  • 61.Thomas SY, et al. Multiple chemokine receptors, including CCR6 and CXCR3, regulate antigen-induced T cell homing to the human asthmatic airway. J Immunol. 2007;179(3):1901–12. doi: 10.4049/jimmunol.179.3.1901. [DOI] [PubMed] [Google Scholar]
  • 62.Rubie C, et al. CCL20/CCR6 expression profile in pancreatic cancer. J Transl Med. 2010;8:45. doi: 10.1186/1479-5876-8-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Matsui T, et al. Selective recruitment of CCR6-expressing cells by increased production of MIP-3 alpha in rheumatoid arthritis. Clin Exp Immunol. 2001;125(1):155–61. doi: 10.1046/j.1365-2249.2001.01542.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Murakami M, et al. Local microbleeding facilitates IL-6- and IL-17-dependent arthritis in the absence of tissue antigen recognition by activated T cells. J Exp Med. 2011;208(1):103–14. doi: 10.1084/jem.20100900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gosselin A, et al. Peripheral blood CCR4+CCR6+ and CXCR3+CCR6+CD4+ T cells are highly permissive to HIV-1 infection. J Immunol. 2010;184(3):1604–16. doi: 10.4049/jimmunol.0903058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Weissman D, et al. Both a precursor and a mature population of dendritic cells can bind HIV. However, only the mature population that expresses CD80 can pass infection to unstimulated CD4+ T cells. J Immunol. 1995;155(8):4111–7. [PubMed] [Google Scholar]
  • 67.Ghosh M, et al. CCL20/MIP3alpha is a novel anti-HIV-1 molecule of the human female reproductive tract. Am J Reprod Immunol. 2009;62(1):60–71. doi: 10.1111/j.1600-0897.2009.00713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fontaine J, Poudrier J, Roger M. Short communication: persistence of high blood levels of the chemokines CCL2, CCL19, and CCL20 during the course of HIV infection. AIDS Res Hum Retroviruses. 2011;27(6):655–7. doi: 10.1089/aid.2010.0261. [DOI] [PubMed] [Google Scholar]
  • 69.Scapini P, et al. Neutrophils produce biologically active macrophage inflammatory protein-3alpha (MIP-3alpha)/CCL20 and MIP-3beta/CCL19. Eur J Immunol. 2001;31(7):1981–8. doi: 10.1002/1521-4141(200107)31:7&#60;1981::aid-immu1981&#62;3.0.co;2-x. [DOI] [PubMed] [Google Scholar]
  • 70.Dieu-Nosjean MC, et al. Regulation of dendritic cell trafficking: a process that involves the participation of selective chemokines. J Leukoc Biol. 1999;66(2):252–62. doi: 10.1002/jlb.66.2.252. [DOI] [PubMed] [Google Scholar]
  • 71.Darash-Yahana M, et al. The chemokine CXCL16 and its receptor, CXCR6, as markers and promoters of inflammation-associated cancers. PLoS One. 2009;4(8):e6695. doi: 10.1371/journal.pone.0006695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Rutter M, et al. Severity of inflammation is a risk factor for colorectal neoplasia in ulcerative colitis. Gastroenterology. 2004;126(2):451–9. doi: 10.1053/j.gastro.2003.11.010. [DOI] [PubMed] [Google Scholar]
  • 73.Erreni M, et al. Expression of chemokines and chemokine receptors in human colon cancer. Methods Enzymol. 2009;460:105–21. doi: 10.1016/S0076-6879(09)05205-7. [DOI] [PubMed] [Google Scholar]
  • 74.Kulbe H, et al. A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment. Cancer Res. 2011 doi: 10.1158/0008-5472.CAN-11-2178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Schutyser E, et al. Identification of biologically active chemokine isoforms from ascitic fluid and elevated levels of CCL18/pulmonary and activation-regulated chemokine in ovarian carcinoma. J Biol Chem. 2002;277(27):24584–93. doi: 10.1074/jbc.M112275200. [DOI] [PubMed] [Google Scholar]
  • 76.Brysse A, et al. Regulation of CXCL8/IL-8 expression by Zonula Occludens-1 in human breast cancer cells. Mol Cancer Res. 2011 doi: 10.1158/1541-7786.MCR-11-0180. [DOI] [PubMed] [Google Scholar]
  • 77.Mantovani A, et al. Cancer-related inflammation. Nature. 2008;454(7203):436–44. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
  • 78.Sica A, Allavena P, Mantovani A. Cancer related inflammation: the macrophage connection. Cancer Lett. 2008;267(2):204–15. doi: 10.1016/j.canlet.2008.03.028. [DOI] [PubMed] [Google Scholar]
  • 79.Treilleux I, et al. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res. 2004;10(22):7466–74. doi: 10.1158/1078-0432.CCR-04-0684. [DOI] [PubMed] [Google Scholar]
  • 80.Bonnotte B, et al. MIP-3alpha transfection into a rodent tumor cell line increases intratumoral dendritic cell infiltration but enhances (facilitates) tumor growth and decreases immunogenicity. J Immunol. 2004;173(8):4929–35. doi: 10.4049/jimmunol.173.8.4929. [DOI] [PubMed] [Google Scholar]
  • 81.Gabrilovich DI, Ciernik IF, Carbone DP. Dendritic cells in antitumor immune responses. I. Defective antigen presentation in tumor-bearing hosts. Cell Immunol. 1996;170(1):101–10. doi: 10.1006/cimm.1996.0139. [DOI] [PubMed] [Google Scholar]
  • 82.Almand B, et al. Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res. 2000;6(5):1755–66. [PubMed] [Google Scholar]
  • 83.Le Mercier I, et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res. 2013;73(15):4629–40. doi: 10.1158/0008-5472.CAN-12-3058. [DOI] [PubMed] [Google Scholar]
  • 84.Marsigliante S, Vetrugno C, Muscella A. CCL20 induces migration and proliferation on breast epithelial cells. J Cell Physiol. 2013;228(9):1873–83. doi: 10.1002/jcp.24349. [DOI] [PubMed] [Google Scholar]
  • 85.Sheng S, Qiao M, Pardee AB. Metastasis and AKT activation. J Cell Physiol. 2009;218(3):451–4. doi: 10.1002/jcp.21616. [DOI] [PubMed] [Google Scholar]
  • 86.Krueger JS, et al. Temporal and quantitative regulation of mitogen-activated protein kinase (MAPK) modulates cell motility and invasion. Oncogene. 2001;20(31):4209–18. doi: 10.1038/sj.onc.1204541. [DOI] [PubMed] [Google Scholar]
  • 87.Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39–51. doi: 10.1016/j.cell.2010.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4(1):71–8. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
  • 89.Levano KS, Jung EH, Kenny PA. Breast cancer subtypes express distinct receptor repertoires for tumor-associated macrophage derived cytokines. Biochem Biophys Res Commun. 2011;411(1):107–10. doi: 10.1016/j.bbrc.2011.06.102. [DOI] [PubMed] [Google Scholar]
  • 90.Sharma M, et al. Analysis of stromal signatures in the tumor microenvironment of ductal carcinoma in situ. Breast Cancer Res Treat. 2010;123(2):397–404. doi: 10.1007/s10549-009-0654-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ojalvo LS, et al. High-density gene expression analysis of tumor-associated macrophages from mouse mammary tumors. Am J Pathol. 2009;174(3):1048–64. doi: 10.2353/ajpath.2009.080676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bierie B, et al. Transforming growth factor-beta regulates mammary carcinoma cell survival and interaction with the adjacent microenvironment. Cancer Res. 2008;68(6):1809–19. doi: 10.1158/0008-5472.CAN-07-5597. [DOI] [PubMed] [Google Scholar]
  • 93.Mohamed MM, Sloane BF. Cysteine cathepsins: multifunctional enzymes in cancer. Nat Rev Cancer. 2006;6(10):764–75. doi: 10.1038/nrc1949. [DOI] [PubMed] [Google Scholar]
  • 94.Verollet C, et al. Extracellular proteolysis in macrophage migration: losing grip for a breakthrough. Eur J Immunol. 2011;41(10):2805–13. doi: 10.1002/eji.201141538. [DOI] [PubMed] [Google Scholar]
  • 95.Hasan L, et al. Function of liver activation-regulated chemokine/CC chemokine ligand 20 is differently affected by cathepsin B and cathepsin D processing. J Immunol. 2006;176(11):6512–22. doi: 10.4049/jimmunol.176.11.6512. [DOI] [PubMed] [Google Scholar]
  • 96.Glondu M, et al. A mutated cathepsin-D devoid of its catalytic activity stimulates the growth of cancer cells. Oncogene. 2001;20(47):6920–9. doi: 10.1038/sj.onc.1204843. [DOI] [PubMed] [Google Scholar]
  • 97.Wolf M, et al. Cathepsin D specifically cleaves the chemokines macrophage inflammatory protein-1 alpha, macrophage inflammatory protein-1 beta, and SLC that are expressed in human breast cancer. Am J Pathol. 2003;162(4):1183–90. doi: 10.1016/s0002-9440(10)63914-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sloane BF, et al. Cathepsin B and tumor proteolysis: contribution of the tumor microenvironment. Semin Cancer Biol. 2005;15(2):149–57. doi: 10.1016/j.semcancer.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 99.Victor BC, et al. Inhibition of cathepsin B activity attenuates extracellular matrix degradation and inflammatory breast cancer invasion. Breast Cancer Res. 2011;13(6):R115. doi: 10.1186/bcr3058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cavallo-Medved D, Moin K, Sloane BF. Cathepsin B. UCSD Nature Molecule Pages. 2011 [PMC free article] [PubMed] [Google Scholar]
  • 101.Chen K, Rajewsky N. The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 2007;8(2):93–103. doi: 10.1038/nrg1990. [DOI] [PubMed] [Google Scholar]
  • 102.Iorio MV, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65(16):7065–70. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
  • 103.Iorio MV, Croce CM. MicroRNAs in cancer: small molecules with a huge impact. J Clin Oncol. 2009;27(34):5848–56. doi: 10.1200/JCO.2009.24.0317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Iyevleva AG, et al. High level of miR-21, miR-10b, and miR-31 expression in bilateral vs. unilateral breast carcinomas. Breast Cancer Res Treat. 2011 doi: 10.1007/s10549-011-1845-z. [DOI] [PubMed] [Google Scholar]
  • 105.Yao T, Lin Z. MiR-21 is involved in cervical squamous cell tumorigenesis and regulates CCL20. Biochim Biophys Acta. 2011 doi: 10.1016/j.bbadis.2011.09.018. [DOI] [PubMed] [Google Scholar]
  • 106.Terao M, et al. Induction of miR-21 by retinoic acid in estrogen receptor-positive breast carcinoma cells: biological correlates and molecular targets. J Biol Chem. 2011;286(5):4027–42. doi: 10.1074/jbc.M110.184994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Asiedu MK, et al. TGFbeta/TNF(alpha)-mediated epithelial-mesenchymal transition generates breast cancer stem cells with a claudin-low phenotype. Cancer Res. 2011;71(13):4707–19. doi: 10.1158/0008-5472.CAN-10-4554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Xu L, et al. Enrichment of CCR6+Foxp3+ regulatory T cells in the tumor mass correlates with impaired CD8+ T cell function and poor prognosis of breast cancer. Clin Immunol. 2010;135(3):466–75. doi: 10.1016/j.clim.2010.01.014. [DOI] [PubMed] [Google Scholar]

RESOURCES