Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 8.
Published in final edited form as: Surgery. 2014 Feb 8;155(6):961–973. doi: 10.1016/j.surg.2014.02.006

Chemokines and chemokine receptors: Update on utility and challenges for the clinician

Ishan Roy a, Douglas B Evans b, Michael B Dwinell a
PMCID: PMC4390364  NIHMSID: NIHMS672699  PMID: 24856117

Chemokines, or chemotactic cytokines, represent alarge family of secreted proteins that have a wide range of function in normal physiology. Chemokine functions include direction of immune cell trafficking, angiogenesis, and wound healing, all critically important to patients being considered for surgery or anticancer therapy. In recent years, perturbations in expression of chemokines or their cognate receptors have been associated with several inflammatory disorders and both solid-tumor and hematologic malignancies. As such, chemokines and chemokine receptors have made attractive targets for biomarker identification and drug discovery. To move chemokine laboratory science to the bedside application for biomarker and drug discovery, we will need a more detailed understanding of the mechanisms by which chemokines are expressed and function. Specific areas that require further study include the structure-function relationship of chemokines and their receptors, differential signaling of ligands, concentration gradient-dependent signaling, and the genetic or epigenetic mechanisms that regulate chemokine ligand or receptor gene expression.

Chemokines provide a powerful opportunity for translational investigation. Chemokines are secreted proteins that travel in the circulation, move through the parenchyma and extracellular matrix of tissues, and bind to and activate the extracellular domain of their cognate receptors present on individual cell types. Consequently, chemokines can serve both as potential biomarkers and as promising targets for pharmaceutical intervention. Similar to other cytokines, chemokines altered in expression during specific disease states can serve as useful diagnostic or prognostic biomarkers. For example, chemokines such as CCL2, CCL5, and CCL20 are potential candidate biomarkers in atherosclerosis, diabetes, and inflammatory diseases of the skin and gut. Herein, we outline the numerous inflammatory diseases and cancers associated with aberrant chemokine expression and activity (Table). Our laboratory is currently investigating the role of several chemokines in pancreatic cancer biology, where inflammation is a key component of tumor initiation, growth, and possibly metastasis. We also discuss the existing opportunities for pharmaceutic targeting of aberrant chemokine activity to alleviate disease. In particular, the chemokine CXCL12 is a potential target for pharmaceutic intervention in several malignancies, including colorectal, breast, and lung cancer. Finally, we explore the emerging pharmaceutic strategies to target more specifically chemokine signaling, leading to a more favorable therapeutic profile.

Table.

Changes in chemokine ligand and receptor expression in human disease

Chemokine ligand Chemokine receptor(s) Human disease/ condition associated Receptor change Ligand change Clinical finding
CCL1 CCR8
CCL2 CCR2, CCR4 Atherosclerosis, type 2 diabetes ↑CCL2 ↑ chemokine levels in plaques, adipose tissue, and circulation leading to macrophage recruitment76,79
CCL3 CCR2, CCR4, CCR5
CCL4 CCR5, CCR8?
CCL5 CCR1, CCR3, CCR4, CCR5 Atherosclerosis, multiple sclerosis ↑CCR1, ↑CCR5 ↑CCL5 Inflammatory chemokine production leading to lymphocytic and monocytic infiltration. Infiltrating monocytes have ↑ CCR1 and ↑ CCR574,77,78
CCL6 CCR1, CCR2, CCR3
CCL7 CCR1, CCR2, CCR3
CCL8 CCR1, CCR2, CCR3
CCL11 CCR3
CCL12 CCR2
CCL13 CCR2, CCR3
CCL14 CCR1
CCL15 CCR1, CCR3
CCL16 CCR1
CCL17 CCR4 Psoriasis, atopic dermatitis ↑CCL17 Keratinocytic/endothelial chemokine production leading to lymphocytic infiltration71,73
CCL18
CCL19 CCR7 Colitis ↑CCL19 Mucosal CCL19 production leading to CCR7-dependent lymphocyte cell infiltration64
CCL20 CCR6 Psoriasis ↑CCL20 Epithelial CCL20 production leading to T-cell infiltration72
Colorectal cancer ↑CCR6 ? ↑ tumor expression of receptor correlated with liver metastasis93
Colitis ↑CCL20 Mucosal CCL20 production leading to CCR6-dependent dendritic cell infiltration22
CCL21 CCR7 Colitis ↑CCL21 Mucosal CCL21 production leading to CCR7-dependent lymphocyte cell infiltration64
Melanoma, gastric cancer, nonsmall cell lung cancer, esophageal cancer, chronic lymphocytic leukemia ↑CCR7 ? Tumor cell expression of CCR7 leads to lymph node metastasis115
CCL22 CCR4 Psoriasis ↑CCL22 Keratinocytic/endothelial chemokine production leading to lymphocytic infiltration71
CCL23 CCR1
CCL24 CCR3
CCL25 CCR9 Colitis ↑CCL25 Mucosal chemokine production leading to lymphocyte cell infiltration65
CCL26 CCR3
CCL27 CCR10 Dermatitis ↑CCL27 Keratinocytic CCL27 production leading to T-cell infiltration70
CCL28 CCR3, CCR10 Colitis ↑CCL28 Mucosal chemokine production leading to lymphocyte cell infiltration66
CXCL1 CXCR1, CXCR2
CXCL2 CXCR2
CXCL3 CXCL4 CXCR2
CXCL5 CXCR2
CXCL6 CXCR1, CXCR2
CXCL7 CXCR1, CXCR2
CXCL8 CXCR1, CXCR2 Colitis ↑CXCL8 Mucosal chemokine production leading to leukocyte cell infiltration116
CXCL9 CXCR3 Psoriasis, multiple sclerosis ↑CXCL9 Inflammatory chemokine production leading to lymphocytic infiltration71,74
CXCL10 CXCR3 Psoriasis, multiple sclerosis ↑CXCL10 Inflammatory chemokine production leading to lymphocytic infiltration71,74
Hepatitis C ↑CXCL10 Increased chemokine levels in sera correlates with poor response to standard treatment4
CXCL11 CXCR3, CXCR7
CXCL12 CXCR4, CXCR7 Colorectal cancer, breast cancer, osteosarcoma, lung cancer, pancreatic cancer ↑CXCR4 ↓CXCL12 Parallel ↑ in receptor expression and ↓ in ligand expression in tumor cells lead to ↑ distant metastasis5659,80,88
Melanoma, ovarian cancer, acute lymphoblastic leukeumia, chronic myelogenous leukemia ↑CXCR4 ? ↑ expression of receptor in tumor cells3,4
CXCL13 CXCR5 Breast cancer ↑CXCR5 ↑CXCL13 In HER2-positive patients, ↑ tumoral ligand and receptor expression correlates with ↑ survival87
CXCL14 Lung cancer ↓CXCL14 Chemokine is silenced in tumor cells, correlated with ↓ tumor necrosis57
CXCL15
CXCL16 CXCR6 Colorectal cancer, renal cancer, breast cancer ↑CXCL16 ↑ tumoral ligand secretion (detected in tissue or serum) correlates with ↑ survival or ↑ radiation response9092
Myocardial infarction/angina pectoris ↓CXCL16 Decreased chemokine levels in sera after episodes78
CXCL17
XCL1 XCR1
XCL2 XCR1
CX3CL1 CX3CR1

BIOCHEMISTRY

Relatively small in size (8–14 kDa), chemotactic cytokines are soluble proteins produced and secreted by a variety of cells. The chemokine ligand family currently has 48 characterized members that can be subdivided into 4 groups on the basis of secondary structure involving spacing between conserved cysteines in the N-terminus of the molecule: CC, CXC, CX3C, and C.14 Chemokine ligand expression can be inducible or constitutive, whereas some ligands are expressed under conditions of inflammation in certain cell types but homeostatically expressed in others. CXC chemokines can be subclassified by the presence or absence of a glutamic acid-leucine-arginine (ELR) motif preceding the first amino-terminal cysteine residue.5

Chemokine receptors are composed of seven transmembrane domain–containing G protein–coupled receptors (GPCRs) and are named on the basis of the secondary structure of their respective ligand binding partners.1 Approximately 40 kDa in size, chemokine receptors signal through several G protein subunits, leading to a diverse array of effectors.6 The conventional signaling pathway attributed to chemokine receptors involves the mobilization of calcium from intracellular stores. Release of calcium is rapid and typically sensitive to blockade of the Gαi subunit of the heterotrimeric G protein complex. Additional signaling targets have been detailed, and include, but are not limited to, phospholipase C and monomeric GTPases, the mitogen-activated protein kinase and phosphatidylinositide 3-kinase pathways, and several tyrosine kinase pathways.713

Although several signaling effectors for chemokines have been identified, their exact mechanism of action and the resulting physiologic functions within specific cell types remains ambiguous. Signaling may be cell-type specific, reflect receptor dimerization, or be altered by unique physiologic contexts, including inflammation, tumor microenvironment, embryogenesis, or angiogenesis. Moving forward, several intracellular and extracellular signaling effectors of chemokines are still being uncovered, in particular with respect to varying diseased states, with the use of unbiased approaches. An example of such a discovery-based approach is the recent mass spectrometric examination of total cellular phosphoproteomic changes after CXCL12 stimulation of T cells.14

Chemokines bind to the extracellular components of their cognate receptors, which elicits distinct signaling events dependent on each unique chemokine and receptor pairing.15,16 Unique pairings of ligands and receptors afford chemokines an assortment of physiologic functions. In some cases, ligands such as CCL2 or CXCL8, are capable of binding to several different receptors.17 In other situations, such as with CCL20-CCR6, single ligands bind exclusively to a single receptor.

Regardless of the exclusivity of binding partners in a particular chemokine signaling axis, studies in which the authors examine expression levels of chemokines or receptors must evaluate all members of a particular signaling partnership simultaneously. Because chemokines direct cell function in a concentration-dependent manner, information regarding chemokine expression must be viewed with this understanding. For example, studies revealing expression of a particular chemokine receptor, while disregarding expression status of the receptor’s binding partners, offer little scientific or clinical impact because they may underrepresent a majority of the signaling effects. Thus, any conclusions drawn from such clinical studies examining a single component of a chemokine axis are of limited utility. The delicate balance between available chemokine ligands and cell-surface levels of chemokine receptors is crucial to normal physiologic functions. This veritable ‘‘Goldilocks’’ effect of chemokines---too much or too little chemokine signaling can lead to unintended consequences and exacerbation of disease---can occur as the result of shifts in either ligand or receptor expression. Thus, the complementary nature of chemokine ligands and receptors must be a central consideration for any future studies of chemokine biology and particularly any putative pharmaceutic strategies. Herein, we briefly summarize the known roles of chemokines in human disease pathophysiology and discuss potential clinical implications while also considering joint ligand-receptor relationships.

CHEMOKINE PHYSIOLOGY

Chemokines in immunity

Chemokine-based mediation of cell movement has been characterized in nearly every cell type, although the physiologic function most associated with chemokine stimulation is recruitment of immune cells.15 Different sets of chemokines are capable of directing the trafficking of unique immune cells, both in inflamed and homeostatic conditions. Constitutively produced chemokines are involved in recruitment of immune cells under nonstressed conditions. These homeostatic chemokines function to repopulate tissues that store immune cells, such as lymph nodes or Peyer’s patches.16,18 For example, the chemokine CCL21, expressed highly in endothelial cells in lymph node venules, mediates the emigration of T cells from lymph vessels to secondary lymphoid tissues.16

By contrast, during pathogenic inflammation, such as with infection, trauma, or cancer, production of an alternate set of chemokines may be induced.19 The production of such inducible chemokines typically is regulated by cytokine production in the local environment of inflammation.20 Inflammatory cytokines such as tumor necrosis factor and interferon-γ induce transcription of a wide variety of chemokines, including CCL2, CCL3, CCL4, CCL5, CCL25, CCL27, CXCL9, and CXCL10.21,22 These chemokines then attract activated cells of both the innate and adaptive immune compartments.23 Importantly, chemokines direct the trafficking of immune cells through both the circulation, in either blood, lymph vessels, and tissue, including environment-specific stroma and extracellular matrices. Unchecked expression of several chemokines, including CCL17, CCL19, CCL20, CCL21, CCL22, CCL25, CCL27, and CCL28, is associated with many different autoimmune and inflammatory disorders, such as colitis and dermatitis (Fig, A).22,24

Fig.

Fig

Schematic overview of chemokine function in intestinal epithelium. Pathologically established gradients of chemokine proteins lead to aberrant migration of cells in multiple ways. Herein, we describe the differing roles of chemokines by using the intestinal epithelium as a model tissue site of dysregulated chemokine production. (A) Acute or chronic inflammation resulting from epithelial barrier disruption, pathogen infection, or other stimuli, leads to increased chemokine secretion. As a result, new local chemokine gradients attract immune cells from the circulatory system into to the site of inflammation. (B) A contrasting gradient is seen during human cancer malignancy, specifically in the CXCL12-CXCR4 axis. Dysplastic lesions contain heterogeneous groups of cells, including some that lose expression of homeostatically produced chemokines, such as CXCL12. Within malignant tumors, those cells that lack CXCL12 ligand expression and retain CXCR4 receptor expression are able to migrate toward existing local gradients of chemokine expression. Primary sources of CXCL12 gradients include neighboring fibroblasts and endothelial cells. Thus, cancer cells lacking ligand expression are capable of following a new gradient into circulation. (C) Those cancer cells that lack CXCL12 ligand expression and survive in circulation can then migrate to distant organs with constitutively high CXCL12 expression, such as the liver, lung, or bone. Within the vessels of these distant sites, cancer cells then follow a new chemokine gradient established by both the endothelial and parenchymal cells, engendering metastases.

Chemokines in angiogenesis and embryogenesis

Chemokines are well characterized in their ability to regulate angiogenesis.2527 Although not absolute, the presence or absence of the ELR motif in CXC chemokines correlates with the ability to be either angiogenic or angiostatic, respectively.28 ELR motif–containing angiogenic chemokines include CXCL1-3 and CXCL5-8, which bind to CXCR1 and CXCR2. Direction of individual events of angiogenesis by these chemokines is partially facilitated by differential binding to each receptor.27 For example, although several ligands can engage both receptors, only ligand binding to CXCR2 leads to endothelial cell migration.29 In addition, the CXCL12-CXCR4 ligand-receptor pair is an independent axis that is angiogenic.26,30 Angiostatic chemokines include CXCL4, CXCL9, CXCL10, and CXCL11. CXCL9, CXCL10, and CXCL11 play additional roles outside of angiogenesis, including T-cell chemoattraction via CXCR3 binding.31 The balance of available proangiogenic ELR+ and angiostatic ELR+ chemokines regulates several functions of endothelial cells, including migration, invasion, proliferation, and endothelial tube formation.26,27

Chemokines clearly play a role in neovascularization during embryogenesis, such as vasculariza-ton of the gut through the CXCL12-CXCR4 axis, but the functions of chemokines during embryonic development are not limited to the vasculature.32 The CXCL12–CXCR4–CXCR7 axis has been implicated in the development of hematopoetic cells, neurogenesis, cardiogenesis, stem cell homing, receptor-specific primordial tissue organization, and endodermal to mesenchymal cell-cell communication.3341 More recently, CX3CL1, CCL14, and CCL4 have been implicated in fetomaternal interactions by directing trophoblast migration along maternal endometria.42

Chemokines in wound healing

Wound healing represents a complex reaction,42 typically involving the epithelial barrier of a given organ surface. Chemokines play an integral part in directing wound closure and healing. Processes such as angiogenesis and recruitment of immune cells, outlined previously, require chemokine stimulation and are similarly involved in wound healing and the response to inflammation.4345 Epithelial cells also express a wide variety of both chemokine ligands and receptors involved in several steps in the reconstitution of the epithelial barrier. Epithelial cell functions regulated by chemokines during wound healing include cell migration, extracellular matrix deposition, cellular adhesion, and cell proliferation.7,46,47 Chemokines, such as CXCL12, CXCL1, and CXCL8, guide these steps of re-epithelialization in tissues such as the gut and skin.47,48 As mentioned previously, unchecked expression of inducible chemokines normally involved in immune cell trafficking can lead to chronic inflammatory states, such as is the case with colitis or dermatitis. Similarly, the dysregulated expression of chemokines specifically involved in epithelial wound healing can lead to pathologic states, as occurs with CXCL12 expression in patients with inflammatory bowel disease (Fig, A).49 The current theory is that abnormally high levels of CXCL12 disrupts the balance in function of the chemokine, perhaps shifting the ligand from epithelial-centric functions in intestinal epithelial cell migration and/or maturation during wound healing to stimulating recruitment of immune cells to the lamina propria (Fig, A).47,49

Emerging concepts in chemokine biology

As seen in the processes of wound healing, chemokines direct not only the act of cell movement, but many other cellular functions, such as proliferation and cell-cell or cell-matrix adhesion. Chemokines regulate the entire process of cellular trafficking into tissues, including intravasation, gradient-dependent migration, cell-cell adhesion, and extravasation. During cell migration, chemokine receptor binding leads to signaling events that cause changes in the cell machinery that facilitate directed cell movement.7 When cells near their destinations and begin interactions with the endothelium, chemokines regulate the transitions that take place to allow the migrating cell to interact with the vascular surface and undergo transendothelial migration.50 For example, the shift from selectin-mediated endothelial interaction to integrin-mediated adhesion required during the extravasation process is mediated by chemokines.51,52 The mechanisms behind the ability of chemokines to regulate multiple distinct cellular functions, such as migration and adhesion or proliferation and apoptosis, have yet to be fully explored. The observation that chemokines stimulate migration in a biphasic, concentration-dependent manner is an important first step in understanding how chemokines are able to dually regulate cell function.53 CXC chemokines such as CXCL12 have an equilibrium concentration for stimulating migration, beyond which the oligomeric chemokine does not stimulate migration.54 It is, therefore, possible that ligand concentration gradients play key regulatory roles in the ability of chemokines to stimulate both migratory and nonmigratory functions.

Specific chemokine axes provide unique opportunities to examine chemokine-driven nonmigratory functions. For example, the functional significance of the near-ubiquitous expression of CXCL12 in normal epithelial cells was previously unknown.55 More recent studies suggest that CXCL12 expression may serve as a checkpoint for preventing malignancy; expression of the ligand is a natural stop-gap that is epigenetically silenced in cancer progression and metastasis from primary tissues such as colon, breast, lung, or bone (Fig, B and C).5659 In particular, re-establishment of CXCL12 expression in colorectal cancer cell lines leads to detachment-based apoptosis or anoikis.8,60 As an additional example, CXCL14 has more direct effects in disease prevention.11,57,61,62 CXCL14 expressed in the epithelium of a variety of tissues has direct antimicrobial effects during bacterial infection as well as the ability to drive necrosis after forced re-expression in lung cancer.57,63 As proteins whose functions occur at the interface between disease and normalcy, CXCL12 and CXCL14 represent unique opportunities for the development of disease-specific biomarkers and pharmaceutic targeting.

CHEMOKINE PATHOPHYSIOLOGY

Chemokines as indicators of disease (biomarkers)

Attention is focusing on the interplay between ligand and receptor expression that influences the ability of individual chemokines to differentially signal. With regard to clinical impact, knowledge of relative concentration levels of particular chemokines may be just as important as determining which chemokines are expressed. Shifts in ligand or receptor expression reflect the basic biology underlying a particular disease. Diseases characterized by local overstimulation of chemokines can lead to pathologies that involve nonspecific localized cell migration, such as is the case with atopic dermatitis. In contrast, pathologically low local chemokine concentrations, such as is the case of CXCL12 in many cancers, could facilitate the emigration of cells from the local environment, potentially through the physiologically established endocrine gradients. These contrasting mechanisms for disease are discussed in more detail herein (Table).

Chemokines in inflammation

Autoimmune diseases are characterized by a state of unchecked inflammation and increased immune cell infiltration into the tissue parenchyma. Diseases such as Crohn’s disease or ulcerative colitis have been shown to have increased expression of the chemokines CCL19, CCL21, CCL25, CCL28, and CXCL8, each of which was correlated with increased mucosal lymphocyte and leukocyte infiltration.6468 Similar examples are found in inflammatory diseases of the skin, with the chemokines CCL17, CCL20, CCL22, CCL27, CXCL9, and CXCL10 thought to regulate the chemoattraction of various dermal infiltrating effector cells.6973 In both psoriasis and atopic dermatitis, excessive recruitment of T cells is driven by chemokines such as CCL17, CCL20, CCL22, and CCL27. In chronic diseases such as type-2 diabetes, multiple sclerosis, and atherosclerosis, increased expression of the chemokines CCL2, CCL5, CXCL9, and CXCL10 correlate with increased infiltration of macrophages and lymphocytes in lesions and plaques.7479 In each of these diseases, permanently increased levels of specific chemokine ligands contribute to ongoing immune cell infiltration.

As summarized by Zlotnik and Yoshie4 in the case of CXCL10, pathologically altered levels of chemokines can serve as useful biomarkers. Hepatitis C patients with increased CXCL10 levels in sera demonstrate poor long-term response to standard treatments.4 In contrast, CXCL16 levels were found to be decreased in the sera of patients after episodes of either angina pectoris or myocardial infarction compared with healthy adults, although no known mechanism has been proposed for this cardioprotection.78

Chemokines in cancer

In contrast, in diseases such as cancer, increased expression of chemokines may lead to immune cell filtration that may correlate with increased survival. After the seminal paper by Müller et al 80 that drew attention to the connection between chemokines and cancer, subsequent reports have focused on the role of chemokine receptors in cancer progression while largely ignoring the expression profile of the chemokine ligands that must bind to those receptors to allow for downstream signaling. The paradigm established by the report by Muller et al suggested that analyzing chemokine receptor expression identifies malignant cells capable of metastasis. Since then, CXCR4 expression has been correlated with a variety of epithelial cancers, including skin, breast, ovarian, colorectal, and pancreatic cancers, as well hematologic malignancies such as acute lymphoblastic leukemia and chronic myelogenous leukemia.3,81 Clinical correlations focusing solely on CXCR4 receptor expression, however, are mixed, with relatively weak links between increased expression and poor prognosis in breast or colon cancer.8285

Recent studies have begun to analyze chemokine ligand expression in the context of cancer. As described previously, the epithelial expression of CXCL12, the cognate ligand for CXCR4, is suppressed in several cancers. Importantly, CXCL12 expression is correlated with improved clinical outcomes in osteosarcoma and breast cancer.58,8688 Studies using mammary, melanoma, pancreatic, and colorectal cancer cell lines demonstrated that endogenously or exogenously reintroducing CXCL12 prevents tumor cells from homing to sites of common metastasis such as the liver or lung (Fig, B and C).9,56,59,89 Thus, in the case of colorectal cancer, decreased autocrine expression of CXCL12 in malignant cells may allow the cells to follow paracrine (localized) and endocrine (distant) gradients and circumvent a physiologic fail-safe mechanism for cells that become non-adherent (Fig, B and C).

Other examples in which increased chemokine expression is associated with improved clinical outcomes in cancer patients include CXCL13, CXCL14, and CXCL16.57,87,90 Although the mechanisms for the apparent benefits provided by these chemokines are largely unknown, some evidence has been uncovered for CXCL16; increased levels of CXCL16 secreted by tumor cells and detected in either tumor tissue or patient sera lead to increased infiltration of effector T cells in breast, colorectal, and renal cancers.9092 The invading immune cells allow for an antitumor effect, leading to significant tumor regression. In breast cancer in particular, CXCL16 expression also was increased after ionizing radiation, suggesting that the induced chemokine expression facilitates infiltration of cytotoxic T cells and a successful anti-tumor cell immune reaction.92

Increased expression of receptors such as CCR6 and CCR7 correlates with liver and lymph node metastasis in several solid-tumor malignancies, such as colorectal cancer and melanoma, respectively.24,93 The role that their cognate ligands, CCL20 and the pair of CCL19 and CCL21, respectively, may play in tumor cell biology, however, is unknown. Given the importance of gradient concentration in the ability of cells to migrate in a directed fashion, a reasonable hypothesis would be that the cognate ligands for CCR6 and CCR7 are repressed in cancers that have metastasized to liver or lymph nodes (Fig, B and C). Depending on the original expression of the ligands in precancerous cells, further study of these ligands may provide a novel opportunity for discovering biomarkers linked with disease progression.

Future studies may be best designed to analyze multiple chemokine pathways simultaneously.90 Using arrays to analyze expression in either human cell lines or tissue may be a better way to elucidate the specific chemokine genes that are most altered in expression relative to others.

CHEMOKINES AS PHARMACEUTIC TARGETS

Eliciting cell movement can lead to both positive and negative physiologic outcomes. Efforts to target chemokines will encounter the aforementioned ‘‘Goldilocks’’ effect that occurs in chemokine biology. Thus, approaches to targeting chemokine signaling designed to modulate disease should tread carefully between the balance to either enhance or inhibit aberrant chemotaxis.

Targeting chemokine receptors

The conventional model for targeting chemokine signaling stems from chemokine receptors being classified as members of the super family of GPCRs. Although numerous pharmaceutic agonists and antagonists exist for other families of GPCRs, most notably the β-adrenergic receptor family,94 there has been little progress in the movement of inhibitors of chemokine receptors into the clinic in the last 15 years.

The most recognized example for pharmaceutically targeting of chemokine receptor is the small molecule inhibitor AMD3100 (plerixafor). AMD3100 was proposed originally for treatment of patients with HIV, given the ability of the virus to use CXCR4 as a coreceptor for internalization.95 In clinical trials, however, treatment with AMD3100 had poor efficacy and resulted in dose-limiting cardiac side effects.96 Subsequent studies established the ability of AMD3100 to mobilize hematopoetic stem cells because of the drug’s ability to block localized CXCL12 signaling in bone marrow, thus allowing cells to enter the circulation.97 This finding allowed AMD3100 to be proposed for use as part of short-term regimens for autologous stem cell transplantation applied to patients with leukemia or multiple myeloma.98 In the case of AMD3100, the serendipitous consequences of systemically blocking chemokine receptor signaling were of benefit, albeit in a short-term setting, and was unrelated to the originally targeted disease. AMD3100 is one of many examples of attempts to block chemokine signaling through receptor inhibition.99 The only currently approved clinical drug that targets a chemokine receptor with the goal of blocking a specific disease process involves the CCR5 inhibitor drugs. The CCR5 small molecular inhibitor UK-427857 (mara-viroc) has been approved for use as an HIV coreceptor blocker in approved patients whose disease has proven resistant to conventional therapies.100 Beyond the use of small molecule inhibitors, several neutralizing antibodies against chemokine receptors have been tested in early-phase clinical trials,99 including an antibody designed for blocking CCR2 activity in a variety of inflammatory diseases.101

Novel targeting of chemokines ligands

Alternate strategies that target chemokine signaling include generation of inhibitors that antagonize ligand rather than receptor activity. For example, a neutralizing antibody for CXCL10 has shown promise in early-phase trials for the treatment of ulcerative colitis and rheumatoid arthritis.102 With the field in its infancy, several strategies for ligand activity manipulation through small molecules have been formulated.103,104 One such manipulation of strategy stems from the characterization of the sulfotyrosine binding pocket that is conserved in all chemokines.105 Generation of inhibitors to the sulfotyrosine binding pocket lends potential versatility to targeting either individual chemokines or entire subgroups of ligands.

More recent strategies for targeting dysregulated signaling of chemokines in disease include the development of biologic or gene-based chemokine therapies. The rationale behind these forms of therapy is partly to restore physiologic gradient concentrations of chemokines rather than systemically negate signaling through protein inhibition. Introducing chemokines systemically could circumvent side effects that would occur from systemic blockade of chemokine receptor signaling, as occurred with AMD3100. In cases in which disease leads to increased trafficking of cells in circulation, such as colitis, dermatitis, or cancer, recombinant chemokine would confuse the established abnormal gradients and arrest further cell movement.

Gene therapies offer one mechanism for locally reintroducing chemokines to treat human disease. Using vaccine or viral methods, the authors of several studies in mouse models have demonstrated the ability of gene-delivered chemokines to recruit immune cells to ameliorate disease, by the use of CCL21 for melanoma,106 CXCL10 to improve HPC vaccine immune reaction,107 and CXCL12 or CXCR4 to treat heart failure.108,109 A plasmid encoding CXCL12 has shown clinical efficacy in treating patients with heart failure in a phase 1 clinical trial.110

Chemokines as biologic therapies wherein synthesized recombinant chemokines are administered directly to patients is an emerging pharmaceutic field. Preclinical studies in colorectal cancer and melanoma in which authors used several recombinant variants of CXCL12 have demonstrated the ability of biologic-based therapies to prevent tumor metastasis.9,89 The chemokines used in this study included monomeric and dimeric variants of CXCL12. These variants, representing the low and high ends of the chemokine concentration gradients that are believed to control biphasic migration, were able to regulate cancer cell migration in mouse models. The monomeric variant of CXCL12 is cardioprotective in rat models of cardiac ischemia.111 Our model, underscoring the importance of concentration gradients, is supported by recent studies demonstrating that, in mice, dimeric CCL2 prevents CCR2-mediated recruitment of leukocytes,112 and, in primate models, modified variants of CCL5 prevent sexual transmission of HIV.113,114 Given their ability to differentially manipulate chemokine-directed cell migration, future development of variants such as these will lead to more efficient pharmaceutic targeting of chemokine signaling.

In summary, at the clinical level, pharmaceutic targeting of chemokine signaling has yet to mirror the diversity of the β-adrenergic or neurotransmitter fields.94 The conventional approach of antagonizing chemokine receptors has had limited success, underscoring the need for more novel approaches to generating chemokine pharmaceuticals. Chemokine ligand-based therapies, using biologics or small molecules, offer potential for treating diseases while avoiding many of the shortcomings of receptor inhibition.

In conclusion, the last decade has witnessed several key advancements in understanding the mechanisms behind chemokine activity in normal physiology as well as disease. However, critical gaps in knowledge still exist and require further study. The downstream signaling effectors of chemokines are still being defined, particularly when considering differing cell types, differing disease states, and nonmigratory functions. Similarly, the mechanisms behind the ability of cells to respond differently to varying concentrations of the same chemokine are not well understood. Further study of concentration-based signaling and resulting functionality, as well as structure and physiologic function of chemokine ligands, is necessary to fully understand the potential of alternative strategies for generation of chemokine specific pharmaceuticals. The concept of chemokine ligands existing in monomerdimer equilibriums, with each side of the equilibrium differentially influencing cell function, is needed in in vivo settings to understand the relationship between varying biochemical quaternary states of specific chemokine ligands and the transition from health to disease. A greater shift toward the understanding of chemokine ligand pharmacology in vivo is required as opposed to the current paradigm of focusing attention on receptor-based activity alone. From a clinical perspective, expression analysis must include not only all components of a given axis at once but also multiple axes. Evaluating the expression of a single gene or a single axis may preclude the ability to detect the most effective biomarkers in a particular disease and ignore potential functional redundancies. At present, chemokines are poised to make substantive impacts as both biomarkers and the basis for novel pharmaceutic strategies in treating human disease.

Acknowledgments

We thank Dr Sam Hwang, Dr Brian Volkman, and Anthony Getschman for their critical evaluation and suggestions for the manuscript. We also thank Mary Bryson of Bryson Biomedical for her assistance with the figure illustrations. Dr Dwinell is co-founder with Dr Brian Volkman and Dr Francis Peterson of Protein Foundry, LLC (www.proteinfoundry.com), a producer of molecular grade chemokines for use in biomedical research. The authors have no other conflicts of interest to declare.

References

  • 1.Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, et al. International union of pharmacology. XXII. nomenclature for chemokine receptors. Pharmacol Rev. 2000;52:145–76. [PubMed] [Google Scholar]
  • 2.Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. 2000;12:121–7. doi: 10.1016/s1074-7613(00)80165-x. [DOI] [PubMed] [Google Scholar]
  • 3.Zlotnik A. Chemokines and cancer. Int J Cancer. 2006;119:2026–9. doi: 10.1002/ijc.22024. [DOI] [PubMed] [Google Scholar]
  • 4.Zlotnik A, Yoshie O. The chemokine superfamily revisited. Immunity. 2012;36:705–16. doi: 10.1016/j.immuni.2012.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hébert CA, Vitangcol RV, Baker JB. Scanning mutagenesis of interleukin-8 identifies a cluster of residues required for receptor binding. J Biol Chem. 1991;266:18989–94. [PubMed] [Google Scholar]
  • 6.Neptune ER, Bourne HR. Receptors induce chemotaxis by releasing the βγsubunit of gi, not by activating gq or Gs. Proc Natl Acad Sci U S A. 1997;94:14489–94. doi: 10.1073/pnas.94.26.14489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Moyer RA, Wendt MK, Johanesen PA, Turner JR, Dwinell MB. Rho activation regulates CXCL12 chemokine stimulated actin rearrangement and restitution in model intestinal epithelia. Lab Invest. 2007;87:807–17. doi: 10.1038/labinvest.3700595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Drury LJ, Wendt MK, Dwinell MB. CXCL12 chemokine expression and secretion regulates colorectal carcinoma cell anoikis through bim-mediated intrinsic apoptosis. PLoS One. 2010;5:e12895. doi: 10.1371/journal.pone.0012895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Drury LJ, Ziarek JJ, Gravel S, Veldkamp CT, Takekoshi T, Hwang ST, et al. Monomeric and dimeric CXCL12 inhibit metastasis through distinct CXCR4 interactions and signaling pathways. Proc Natl Acad Sci U S A. 2011;108:17655–60. doi: 10.1073/pnas.1101133108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Agle KA, Vongsa RA, Dwinell MB. Calcium mobilization triggered by the chemokine CXCL12 regulates migration in wounded intestinal epithelial monolayers. J Biol Chem. 2010;285:16066–75. doi: 10.1074/jbc.M109.061416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Murakami T, Cardones AR, Finkelstein SE, Restifo NP, Klaunberg BA, Nestle FO, et al. Immune evasion by murine melanoma mediated through CC chemokine receptor-10. J Exp Med. 2003;198:1337–47. doi: 10.1084/jem.20030593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wong M, Uddin S, Majchrzak B, Huynh T, Proudfoot AEI, Platanias LC, et al. RANTES activates Jak2 and Jak3 to regulate engagement of multiple signaling pathways in T cells. J Biol Chem. 2001;276:11427–31. doi: 10.1074/jbc.M010750200. [DOI] [PubMed] [Google Scholar]
  • 13.Mellado M, Rodríguez-Frade JM, Mañes S, Martínez-A C. Chemokine signaling and functional responses: the role of receptor dimerization and TK pathway activation. Annu Rev Immunol. 2001;19:397–421. doi: 10.1146/annurev.immunol.19.1.397. [DOI] [PubMed] [Google Scholar]
  • 14.Wojcechowskyj JA, Lee JY, Seeholzer SH, Doms RW. Quantitative phosphoproteomics of CXCL12 (SDF-1) signaling. PLoS One. 2011;6:e24918. doi: 10.1371/journal.pone.0024918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rollins BJ. Chemokines. Blood. 1997;90:909–28. [PubMed] [Google Scholar]
  • 16.Cyster JG. Chemokines and cell migration in secondary lymphoid organs. Science. 1999;286:2098–102. doi: 10.1126/science.286.5447.2098. [DOI] [PubMed] [Google Scholar]
  • 17.Paoletti S, Petkovic V, Sebastiani S, Danelon MG, Uguccioni M, Gerber BO. A rich chemokine environment strongly enhances leukocyte migration and activities. Blood. 2005;105:3405–12. doi: 10.1182/blood-2004-04-1648. [DOI] [PubMed] [Google Scholar]
  • 18.Förster R, Schubel A, Breitfeld D, Kremmer E, Renner-Müller I, Wolf E, et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999;99:23–33. doi: 10.1016/s0092-8674(00)80059-8. [DOI] [PubMed] [Google Scholar]
  • 19.Mueller SN, Germain RN. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat Rev Immunol. 2009;9:618–29. doi: 10.1038/nri2588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Barker JN, Sarma V, Mitra RS, Dixit VM, Nickoloff BJ. Marked synergism between tumor necrosis factor-alpha and interferon-gamma in regulation of keratinocyte-derived adhesion molecules and chemotactic factors. J Clin Invest. 1990;85:605–8. doi: 10.1172/JCI114481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ogawa N, Ping L, Zhenjun L, Takada Y, Sugai S. Involvement of the interferon-gamma-induced T cell-attracting chemokines, interferon-gamma-inducible 10-kd protein (CXCL10) and monokine induced by interferon-gamma (CXCL9), in the salivary gland lesions of patients with Sjögren’s syndrome. Arthritis Rheum. 2002;46:2730–41. doi: 10.1002/art.10577. [DOI] [PubMed] [Google Scholar]
  • 22.Zimmerman NP, Vongsa RA, Wendt MK, Dwinell MB. Chemokines and chemokine receptors in mucosal homeostasis at the intestinal epithelial barrier in inflammatory bowel disease. Inflamm Bowel Dis. 2008;14:1000–11. doi: 10.1002/ibd.20480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vecchi A, Massimiliano L, Ramponi S, Luini W, Bernasconi S, Bonecchi R, et al. Differential responsiveness to constitutive vs. inducible chemokines of immature and mature mouse dendritic cells. J Leukoc Biol. 1999;66:489–94. doi: 10.1002/jlb.66.3.489. [DOI] [PubMed] [Google Scholar]
  • 24.Murakami T, Cardones AR, Hwang ST. Chemokine receptors and melanoma metastasis. J Dermatol Sci. 2004;36:71–8. doi: 10.1016/j.jdermsci.2004.03.002. [DOI] [PubMed] [Google Scholar]
  • 25.Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE, Burdick MD, et al. CXC chemokines in angiogenesis. J Leukoc Biol. 2000;68:1–8. [PubMed] [Google Scholar]
  • 26.Heidemann J, Ogawa H, Rafiee P, Lügering N, Maaser C, Domschke W, et al. Mucosal angiogenesis regulation by CXCR4 and its ligand CXCL12 expressed by human intestinal microvascular endothelial cells. Am J Physiol. 2004;286:G1059–68. doi: 10.1152/ajpgi.00417.2003. [DOI] [PubMed] [Google Scholar]
  • 27.Heidemann J, Ogawa H, Dwinell MB, Rafiee P, Maaser C, Gockel HR, et al. Angiogenic effects of interleukin 8 (CXCL8) in human intestinal microvascular endothelial cells are mediated by CXCR2. J Biol Chem. 2003;278:8508–15. doi: 10.1074/jbc.M208231200. [DOI] [PubMed] [Google Scholar]
  • 28.Strieter RM, Polverini PJ, Kunkel SL, Arenberg DA, Burdick MD, Kasper J, et al. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J Biol Chem. 1995;270:27348–57. doi: 10.1074/jbc.270.45.27348. [DOI] [PubMed] [Google Scholar]
  • 29.Addison CL, Daniel TO, Burdick MD, Liu H, Ehlert JE, Xue YY, et al. The CXC chemokine receptor 2, CXCR2, is the putative receptor for ELR+ CXC chemokine-induced angiogenic activity. J Immunol. 2000;165:5269–77. doi: 10.4049/jimmunol.165.9.5269. [DOI] [PubMed] [Google Scholar]
  • 30.Salcedo R, Oppenheim JJ. Role of chemokines in angiogenesis: CXCL12/SDF-1 and CXCR4 interaction, a key regulator of endothelial cell responses. Microcirculation. 2003;10:359–70. doi: 10.1038/sj.mn.7800200. [DOI] [PubMed] [Google Scholar]
  • 31.Hannelien V, Karel G, Jo VD, Sofie S. The role of CXC chemokines in the transition of chronic inflammation to esophageal and gastric cancer. Biochim Biophys Acta. 2012;1825:117–29. doi: 10.1016/j.bbcan.2011.10.008. [DOI] [PubMed] [Google Scholar]
  • 32.Tachibana K, Hirota S, Iizasa H, Yoshida H, Kawabata K, Kataoka Y, et al. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature. 1998;393:591–4. doi: 10.1038/31261. [DOI] [PubMed] [Google Scholar]
  • 33.Katsumoto K, Kume S. Endoderm and mesoderm reciprocal signaling mediated by CXCL12 and CXCR4 regulates the migration of angioblasts and establishes the pancreatic fate. Development. 2011;138:1947–55. doi: 10.1242/dev.058719. [DOI] [PubMed] [Google Scholar]
  • 34.Dambly-Chaudiere C, Cubedo N, Ghysen A. Control of cell migration in the development of the posterior lateral line: antagonistic interactions between the chemokine receptors CXCR4 and CXCR7/RDC1. BMC Dev Biol. 2007;7:23. doi: 10.1186/1471-213X-7-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Valentin G, Haas P, Gilmour D. The chemokine SDF1a coordinates tissue migration through the spatially restricted activation of Cxcr7 and Cxcr4b. Curr Biol. 2007;17:1026–31. doi: 10.1016/j.cub.2007.05.020. [DOI] [PubMed] [Google Scholar]
  • 36.Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–8. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
  • 37.Voigt I, Camacho SA, de Boer BA, Lipp M, Förster R, Berek C. CXCR5-deficient mice develop functional germinal centers in the splenic T cell zone. Eur J Immunol. 2000;30:560–7. doi: 10.1002/1521-4141(200002)30:2<560::AID-IMMU560>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
  • 38.Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–64. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
  • 39.Hannan NJ, Salamonsen LA. Role of chemokines in the endometrium and in embryo implantation. Curr Opin Obstet Gynecol. 2007;19:266–72. doi: 10.1097/GCO.0b013e328133885f. [DOI] [PubMed] [Google Scholar]
  • 40.Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa S, Kitamura Y, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996;382:635–8. doi: 10.1038/382635a0. [DOI] [PubMed] [Google Scholar]
  • 41.Zou Y, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998;393:595–9. doi: 10.1038/31269. [DOI] [PubMed] [Google Scholar]
  • 42.Hannan NJ, Jones RL, White CA, Salamonsen LA. The chemokines, CX3CL1, CCL14, and CCL4, promote human trophoblast migration at the fetomaternal interface. Biol Reprod. 2006;74:896–904. doi: 10.1095/biolreprod.105.045518. [DOI] [PubMed] [Google Scholar]
  • 43.Nanney LB, Mueller SG, Bueno R, Peiper SC, Richmond A. Distributions of melanoma growth stimulatory activity of growth-regulated gene and the interleukin-8 receptor B in human wound repair. Am J Pathol. 1995;147:1248–60. [PMC free article] [PubMed] [Google Scholar]
  • 44.Ishida Y, Gao J, Murphy PM. Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function. J Immunol. 2008;180:569–79. doi: 10.4049/jimmunol.180.1.569. [DOI] [PubMed] [Google Scholar]
  • 45.Inokuma D, Abe R, Fujita Y, Sasaki M, Shibaki A, Nakamura H, et al. CTACK/CCL27 accelerates skin regeneration via accumulation of bone marrow-derived keratinocytes. Stem Cells. 2006;24:2810–6. doi: 10.1634/stemcells.2006-0264. [DOI] [PubMed] [Google Scholar]
  • 46.Dwinell MB, Johanesen PA, Smith JM. Immunobiology of epithelial chemokines in the intestinal mucosa. Surgery. 2003;133:601–7. doi: 10.1067/msy.2003.143. [DOI] [PubMed] [Google Scholar]
  • 47.Smith JM, Johanesen PA, Wendt MK, Binion DG, Dwinell MB. CXCL12 activation of CXCR4 regulates mucosal host defense through stimulation of epithelial cell migration and promotion of intestinal barrier integrity. Am J Physiol. 2005;288:G316–26. doi: 10.1152/ajpgi.00208.2004. [DOI] [PubMed] [Google Scholar]
  • 48.Gillitzer R, Goebeler M. Chemokines in cutaneous wound healing. J Leukoc Biol. 2001;69:513–21. [PubMed] [Google Scholar]
  • 49.Dotan I, Werner L, Vigodman S, Weiss S, Brazowski E, Maharshak N, et al. CXCL12 is a constitutive and inflammatory chemokine in the intestinal immune system. Inflamm Bowel Dis. 2010;16:583–92. doi: 10.1002/ibd.21106. [DOI] [PubMed] [Google Scholar]
  • 50.Butcher EC, Picker LJ. Lymphocyte homing and homeostasis. Science. 1996;272:60–7. doi: 10.1126/science.272.5258.60. [DOI] [PubMed] [Google Scholar]
  • 51.Peled A, Kollet O, Ponomaryov T, Petit I, Franitza S, Grabovsky V, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34+ cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000;95:3289–96. [PubMed] [Google Scholar]
  • 52.Luster AD. Chemokines---chemotactic cytokines that mediate inflammation. N Engl J Med. 1998;338:436–45. doi: 10.1056/NEJM199802123380706. [DOI] [PubMed] [Google Scholar]
  • 53.Moghe PV, Nelson RD, Tranquillo RT. Cytokine-stimulated chemotaxis of human neutrophils in a 3-D conjoined fibrin gel assay. J Immunol Methods. 1995;180:193–211. doi: 10.1016/0022-1759(94)00314-m. [DOI] [PubMed] [Google Scholar]
  • 54.Veldkamp CT, Peterson FC, Pelzek AJ, Volkman BF. The monomer-dimer equilibrium of stromal cell-derived factor-1 (CXCL 12) is altered by pH, phosphate, sulfate, and heparin. Protein Sci. 2005;14:1071–81. doi: 10.1110/ps.041219505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Shirozu M, Nakano T, Inazawa J, Tashiro K, Tada H, Shinohara T, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics. 1995;28:495–500. doi: 10.1006/geno.1995.1180. [DOI] [PubMed] [Google Scholar]
  • 56.Wendt MK, Johanesen PA, Kang-Decker N, Binion DG, Shah V, Dwinell MB. Silencing of epithelial CXCL12 expression by DNA hypermethylation promotes colonic carcinoma metastasis. Oncogene. 2006;25:4986–97. doi: 10.1038/sj.onc.1209505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tessema M, Klinge DM, Yingling CM, Do K, Van Neste L, Belinsky SA. Re-expression of CXCL14, a common target for epigenetic silencing in lung cancer, induces tumor necrosis. Oncogene. 2010;29:5159–70. doi: 10.1038/onc.2010.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Baumhoer D, Smida J, Zillmer S, Rosemann M, Atkinson MJ, Nelson PJ, et al. Strong expression of CXCL12 is associated with a favorable outcome in osteosarcoma. Mod Pathol. 2012;25:522–8. doi: 10.1038/modpathol.2011.193. [DOI] [PubMed] [Google Scholar]
  • 59.Wendt MK, Cooper AN, Dwinell MB. Epigenetic silencing of CXCL12 increases the metastatic potential of mammary carcinoma cells. Oncogene. 2008;27:1461–71. doi: 10.1038/sj.onc.1210751. [DOI] [PubMed] [Google Scholar]
  • 60.Wendt MK, Drury LJ, Vongsa RA, Dwinell MB. Constitutive CXCL12 expression induces anoikis in colorectal carcinoma cells. Gastroenterology. 2008;135:508–17. doi: 10.1053/j.gastro.2008.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A, Soler D, et al. DCs metabolize sunlight-induced vitamin D3 to ’program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol. 2007;8:285–93. doi: 10.1038/ni1433. [DOI] [PubMed] [Google Scholar]
  • 62.Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang L, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and treg cells. Nature. 2011;475:226–30. doi: 10.1038/nature10169. [DOI] [PubMed] [Google Scholar]
  • 63.Maerki C, Meuter S, Liebi M, Mühlemann K, Frederick MJ, Yawalkar N, et al. Potent and broad-spectrum antimicrobial activity of CXCL14 suggests an immediate role in skin infections. J Immunol. 2009;182:507–14. doi: 10.4049/jimmunol.182.1.507. [DOI] [PubMed] [Google Scholar]
  • 64.Kawashima D, Oshitani N, Jinno Y, Watanabe K, Nakamura S, Higuchi K, et al. Augmented expression of secondary lymphoid tissue chemokine and EBI1 ligand chemokine in Crohn’s disease. J Clin Pathol. 2005;58:1057–63. doi: 10.1136/jcp.2004.024828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Papadakis KA, Prehn J, Moreno ST, Cheng L, Kouroumalis EA, Deem R, et al. CCR9-positive lymphocytes and thymus-expressed chemokine distinguish small bowel from colonic crohn’s disease. Gastroenterology. 2001;121:246–54. doi: 10.1053/gast.2001.27154. [DOI] [PubMed] [Google Scholar]
  • 66.Ogawa H, Iimura M, Eckmann L, Kagnoff MF. Regulated production of the chemokine CCL28 in human colon epithelium. Am J Physiol. 2004;287:G1062–9. doi: 10.1152/ajpgi.00162.2004. [DOI] [PubMed] [Google Scholar]
  • 67.Middel P, Raddatz D, Gunawan B, Haller F, Radzun H. Increased number of mature dendritic cells in crohn’s disease: evidence for a chemokine mediated retention mechanism. Gut. 2006;55:220–7. doi: 10.1136/gut.2004.063008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Saruta M, Yu QT, Avanesyan A, Fleshner PR, Targan SR, Papadakis KA. Phenotype and effector function of CC chemokine receptor 9-expressing lymphocytes in small intestinal crohn’s disease. J Immunol. 2007;178:3293–300. doi: 10.4049/jimmunol.178.5.3293. [DOI] [PubMed] [Google Scholar]
  • 69.Mabuchi T, Singh TP, Takekoshi T, Jia GF, Wu X, Kao MC, et al. CCR6 is required for epidermal trafficking of gammadelta-T cells in an IL-23-induced model of psoriasi-form dermatitis. J Invest Dermatol. 2013;133:164–71. doi: 10.1038/jid.2012.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Homey B, Alenius H, Muller A, Soto H, Bowman EP, Yuan W, et al. CCL27-CCR10 interactions regulate T cell-mediated skin inflammation. Nat Med. 2002;8:157–65. doi: 10.1038/nm0202-157. [DOI] [PubMed] [Google Scholar]
  • 71.Rottman JB, Smith TL, Ganley KG, Kikuchi T, Krueger JG. Potential role of the chemokine receptors CXCR3, CCR4, and the integrin alphaEbeta7 in the pathogenesis of psoriasis vulgaris. Lab Invest. 2001;81:335–47. doi: 10.1038/labinvest.3780242. [DOI] [PubMed] [Google Scholar]
  • 72.Homey B, Dieu-Nosjean M, Wiesenborn A, Massacrier C, Pin J, Oldham E, et al. Up-regulation of macrophage inflammatory protein-3α/CCL20 and CC chemokine receptor 6 in psoriasis. J Immunol. 2000;164:6621–32. doi: 10.4049/jimmunol.164.12.6621. [DOI] [PubMed] [Google Scholar]
  • 73.Pivarcsi A, Homey B. Chemokine networks in atopic dermatitis: traffic signals of disease. Curr Allergy Asthma Rep. 2005;5:284–90. doi: 10.1007/s11882-005-0068-y. [DOI] [PubMed] [Google Scholar]
  • 74.Sørensen TL, Tani M, Jensen J, Pierce V, Lucchinetti C, Folcik VA, et al. Expression of specific chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. J Clin Invest. 1999;103:807–15. doi: 10.1172/JCI5150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mahad DJ, Trebst C, Kivisäkk P, Staugaitis SM, Tucky B, Wei T, et al. Expression of chemokine receptors CCR1 and CCR5 reflects differential activation of mononuclear phagocytes in pattern II and pattern III multiple sclerosis lesions. J Neuropathol Exp Neurol. 2004;63:262–73. doi: 10.1093/jnen/63.3.262. [DOI] [PubMed] [Google Scholar]
  • 76.Nelken NA, Coughlin SR, Gordon D, Wilcox JN. Monocyte chemoattractant protein-1 in human atheromatous plaques. J Clin Invest. 1991;88:1121–7. doi: 10.1172/JCI115411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Trebst C, Lykke Sørensen T, Kivisäkk P, Cathcart MK, Hesselgesser J, Horuk R, et al. CCR1+/CCR5+ mononuclear phagocytes accumulate in the central nervous system of patients with multiple sclerosis. Am J Pathol. 2001;159:1701–10. doi: 10.1016/s0002-9440(10)63017-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Barlic J, Murphy PM. Chemokine regulation of atherosclerosis. J Leukoc Biol. 2007;82:226–36. doi: 10.1189/jlb.1206761. [DOI] [PubMed] [Google Scholar]
  • 79.Cardona S, Garcia J, Cardona A. The fine balance of chemokines during disease: trafficking, inflammation, and homeostasis. Methods Mol Biol. 2013;1013:1–16. doi: 10.1007/978-1-62703-426-5_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410:50–6. doi: 10.1038/35065016. [DOI] [PubMed] [Google Scholar]
  • 81.Zlotnik A. Involvement of chemokine receptors in organ-specific metastasis. Contrib Microbiol. 2006;13:191–9. doi: 10.1159/000092973. [DOI] [PubMed] [Google Scholar]
  • 82.Mirisola V, Zuccarino A, Bachmeier BE, Sormani MP, Falter J, Nerlich A, et al. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur J Cancer. 2009;45:2579–87. doi: 10.1016/j.ejca.2009.06.026. [DOI] [PubMed] [Google Scholar]
  • 83.Schimanski CC, Schwald S, Simiantonaki N, Jayasinghe C, Gönner U, Wilsberg V, et al. Effect of chemokine receptors CXCR4 and CCR7 on the metastatic behavior of human colorectal cancer. Clin Cancer Res. 2005;11:1743–50. doi: 10.1158/1078-0432.CCR-04-1195. [DOI] [PubMed] [Google Scholar]
  • 84.Andre F, Xia W, Conforti R, Wei Y, Boulet T, Tomasic G, et al. CXCR4 expression in early breast cancer and risk of distant recurrence. Oncologist. 2009;14:1182–8. doi: 10.1634/theoncologist.2009-0161. [DOI] [PubMed] [Google Scholar]
  • 85.Hassan S, Ferrario C, Saragovi U, Quenneville L, Gaboury L, Baccarelli A, et al. The influence of tumor-host interactions in the stromal cell-derived factor-1/CXCR4 ligand/receptor axis in determining metastatic risk in breast cancer. Am J Pathol. 2009;175:66–73. doi: 10.2353/ajpath.2009.080948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ramos E, Camargo A, Braun K, Slowik R, Cavalli I, Ribeiro E, et al. Simultaneous CXCL12 and ESR1 CpG island hypermethylation correlates with poor prognosis in sporadic breast cancer. BMC Cancer. 2010;10:23. doi: 10.1186/1471-2407-10-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Razis E, Kalogeras KT, Kotoula V, Eleftheraki AG, Nikitas N, Kronenwett R, et al. Improved outcome of high-risk early HER2 positive breast cancer with high CXCL13-CXCR5 messenger RNA expression. Clin Breast Cancer. 2012;12:183–93. doi: 10.1016/j.clbc.2012.03.006. [DOI] [PubMed] [Google Scholar]
  • 88.Roy I, Zimmerman NP, Mackinnon AC, Tsai S, Evans DB, Dwinnell MB. CXCL12 chemokine expression suppresses human pancreatic cancer growth and metastasis. PLoSOne. 2014;9(3):e90400. doi: 10.1371/pone.0090400. Epub. 2014 Mar 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Takekoshi T, Ziarek JJ, Volkman BF, Hwang ST. A locked, dimeric CXCL12 variant effectively inhibits pulmonary metastasis of CXCR4-expressing melanoma cells due to enhanced serum stability. Mol Cancer Ther. 2012;11:2516–25. doi: 10.1158/1535-7163.MCT-12-0494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hojo S, Koizumi K, Tsuneyama K, Arita Y, Cui Z, Shinohara K, et al. High-level expression of chemokine CXCL16 by tumor cells correlates with a good prognosis and increased tumor-infiltrating lymphocytes in colo-rectal cancer. Cancer Res. 2007;67:4725–31. doi: 10.1158/0008-5472.CAN-06-3424. [DOI] [PubMed] [Google Scholar]
  • 91.Gutwein P, Schramme A, Sinke N, Abdel-Bakky MS, Voss B, Obermüller N, et al. Tumoural CXCL16 expression is a novel prognostic marker of longer survival times in renal cell cancer patients. Eur J Cancer. 2009;45:478–89. doi: 10.1016/j.ejca.2008.10.023. [DOI] [PubMed] [Google Scholar]
  • 92.Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M, Cameron TO, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008;181:3099–107. doi: 10.4049/jimmunol.181.5.3099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ghadjar P, Rubie C, Aebersold DM, Keilholz U. The chemokine CCL20 and its receptor CCR6 in human malignancy with focus on colorectal cancer. Int J Cancer. 2009;125:741–5. doi: 10.1002/ijc.24468. [DOI] [PubMed] [Google Scholar]
  • 94.Whalen EJ, Rajagopal S, Lefkowitz RJ. Therapeutic potential of β-arrestin- and G protein-biased agonists. Trends Mol Med. 2011;17:126–39. doi: 10.1016/j.molmed.2010.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Donzella GA, Schols D, Lin SW, Este JA, Nagashima KA, Maddon PJ, et al. AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor. Nat Med. 1998;4:72–7. doi: 10.1038/nm0198-072. [DOI] [PubMed] [Google Scholar]
  • 96.Hendrix CW, Collier AC, Lederman MM, Schols D, Pollard RB, Brown S, et al. Safety, pharmacokinetics, and antiviral activity of AMD3100, a selective CXCR4 receptor inhibitor, in HIV-1 infection. J Acquir Immune Defic Syndr. 2004;37:1253–62. doi: 10.1097/01.qai.0000137371.80695.ef. [DOI] [PubMed] [Google Scholar]
  • 97.Hendrix CW, Flexner C, MacFarland RT, Giandomenico C, Fuchs EJ, Redpath E, et al. Pharmacokinetics and safety of AMD-3100, a novel antagonist of the CXCR-4 chemokine receptor, in human volunteers. Antimicrob Agents Chemother. 2000;44:1667–73. doi: 10.1128/aac.44.6.1667-1673.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Plerixafor: AMD AMD3100, JM 3100, SDZ SID. Drugs R D. 2007;8:113–9. doi: 10.2165/00126839-200708020-00006. [DOI] [PubMed] [Google Scholar]
  • 99.Horuk R. Chemokine receptor antagonists: overcoming developmental hurdles. Nat Rev Drug Discov. 2009;8:23–33. doi: 10.1038/nrd2734. [DOI] [PubMed] [Google Scholar]
  • 100.Abel S, Back DJ, Vourvahis M. Maraviroc: pharmacokinetics and drug interactions. Antivir Ther. 2009;14:607–18. [PubMed] [Google Scholar]
  • 101.Rutgeerts P, Vermeire S, Van Assche G. Biological therapies for inflammatory bowel diseases. Gastroenterology. 2009;136:1182–97. doi: 10.1053/j.gastro.2009.02.001. [DOI] [PubMed] [Google Scholar]
  • 102.Yellin M, Paliienko I, Balanescu A, Ter-Vartanian S, Tseluyko V, Xu L, et al. A phase II, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of MDX-1100, a fully human anti-CXCL10 monoclonal antibody, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheum. 2012;64:1730–9. doi: 10.1002/art.34330. [DOI] [PubMed] [Google Scholar]
  • 103.Galzi J, Hachet-Haas M, Bonnet D, Daubeuf F, Lecat S, Hibert M, et al. Neutralizing endogenous chemokines with small molecules: principles and potential therapeutic applications. Pharmacol Ther. 2010;126:39–55. doi: 10.1016/j.pharmthera.2009.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Daubeuf F, Hachet-Haas M, Gizzi P, Gasparik V, Bonnet D, Utard V, et al. An antedrug of the CXCL12 neutraligand blocks experimental allergic asthma without systemic effect in mice. J Biol Chem. 2013;288:11865–76. doi: 10.1074/jbc.M112.449348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Veldkamp CT, Seibert C, Peterson FC, De la Cruz NB, Haugner JC, 3rd, Basnet H, et al. Structural basis of CXCR4 sulfotyrosine recognition by the chemokine SDF-1/CXCL12. Sci Signal. 2008;1:ra4. doi: 10.1126/scisignal.1160755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Igoucheva O, Grazzini M, Pidich A, Kemp DM, Larijani M, Farber M, et al. Immunotargeting and eradication of orthotopic melanoma using a chemokine-enhanced DNA vaccine. Gene Ther. 2013;20:939–48. doi: 10.1038/gt.2013.17. [DOI] [PubMed] [Google Scholar]
  • 107.Mohit E, Bolhassani A, Zahedifard F, Seyed N, Eslamifar A, Taghikhani M, et al. Immunomodulatory effects of IP-10 chemokine along with PEI600-tat delivery system in DNA vaccination against HPV infections. Mol Immunol. 2013;53:149–60. doi: 10.1016/j.molimm.2012.07.011. [DOI] [PubMed] [Google Scholar]
  • 108.Sundararaman S, Miller TJ, Pastore JM, Kiedrowski M, Aras R, Penn MS. Plasmid-based transient human stromal cell-derived factor-1 gene transfer improves cardiac function in chronic heart failure. Gene Ther. 2011;18:867–73. doi: 10.1038/gt.2011.18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.LaRocca TJ, Jeong D, Kohlbrenner E, Lee A, Chen J, Hajjar RJ, et al. CXCR4 gene transfer prevents pressure overload induced heart failure. J Mol Cell Cardiol. 2012;53:223–32. doi: 10.1016/j.yjmcc.2012.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Penn MS, Mendelsohn FO, Schaer GL, Sherman W, Farr M, Pastore J, et al. An open-label dose escalation study to evaluate the safety of administration of nonviral stromal cell-derived factor-1 plasmid to treat symptomatic ischemic heart failure. Circ Res. 2013;112:816–25. doi: 10.1161/CIRCRESAHA.111.300440. [DOI] [PubMed] [Google Scholar]
  • 111.Ziarek JJ, Veldkamp CT, Zhang F, Murray NJ, Kartz GA, Liang X, et al. Heparin oligosaccharides inhibit chemokine (CXC motif) ligand 12 (CXCL12) cardioprotection by binding orthogonal to the dimerization interface, promoting oligomerization, and competing with the chemokine (CXC motif) receptor 4 (CXCR4) N terminus. J Biol Chem. 2013;288:737–46. doi: 10.1074/jbc.M112.394064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Tan JHY, Ludeman JP, Wedderburn J, Canals M, Hall P, Butler SJ, et al. Tyrosine sulfation of chemokine receptor CCR2 enhances interactions with both monomeric and dimeric forms of the chemokine monocyte chemoattractant protein-1 (MCP-1) J Biol Chem. 2013;288:10024–34. doi: 10.1074/jbc.M112.447359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Simmons G, Clapham PR, Picard L, Offord RE, Rosenkilde MM, Schwartz TW, et al. Potent inhibition of HIV-1 infectivity in macrophages and lymphocytes by a novel CCR5 antagonist. Science. 1997;276:276–9. doi: 10.1126/science.276.5310.276. [DOI] [PubMed] [Google Scholar]
  • 114.Proudfoot AEI, Power CA, Hoogewerf AJ, Montjovent MO, Borlat F, Offord RE, et al. Extension of recombinant human RANTES by the retention of the initiating methionine produces a potent antagonist. J Biol Chem. 1996;271:2599–603. doi: 10.1074/jbc.271.5.2599. [DOI] [PubMed] [Google Scholar]
  • 115.Kakinuma T, Hwang ST. Chemokines, chemokine receptors, and cancer metastasis. J Leukoc Biol. 2006;79:639–51. doi: 10.1189/jlb.1105633. [DOI] [PubMed] [Google Scholar]
  • 116.Fonseca-Camarillo G, Yamamoto-Furusho JK. High gene expression of CXCL8 is associated with the presence of extraintestinal manifestations and long-term disease in patients with ulcerative colitis. Inflamm Bowel Dis. 2013;19:E22–3. doi: 10.1002/ibd.22857. [DOI] [PubMed] [Google Scholar]

RESOURCES