Immunology Letters 145 (2012) 23–29
Contents lists available at SciVerse ScienceDirect
Immunology Letters journal homepage: www.elsevier.com/locate/immlet
Review
Chemokine receptor oligomerization: A further step toward chemokine function ˜ Laura Martínez Munoz, Borja López Holgado, Carlos Martínez-A, José Miguel Rodríguez-Frade, Mario Mellado ∗ Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, Madrid E-28049, Spain
a r t i c l e
i n f o
a b s t r a c t
Keywords: Chemokine receptors Oligomerization GPCR
A broad array of biological responses including cell polarization, movement, immune and inflammatory responses, as well as prevention of HIV-1 infection, are triggered by the chemokines, a family of secreted and structurally related chemoattractant proteins that bind to class A-specific seven-transmembrane receptors linked to G proteins. Chemokines and their receptors should not be considered isolated entities, as they act in complex networks. Chemokines bind as oligomers, or oligomerize after binding to glycosaminoglycans on endothelial cells, and are then presented to their receptors on target cells, facilitating the generation of chemoattractant gradients. The chemokine receptors form homo- and heterodimers, as well as higher order structures at the cell surface. These structures are dynamic and are regulated by receptor expression and ligand levels. Complexity is even greater, as in addition to regulation by cytokines and decoy receptors, chemokine and receptor levels are affected by proteolytic cleavage and other protein modifications. This complex scenario should be considered when analyzing chemokine biology and the ability of their antagonists to act in vivo. Strategies based on blocking or stabilizing ligand and receptor dimers could be alternative approaches that might have broad therapeutic potential. © 2012 Elsevier B.V. All rights reserved.
1. Introduction
Lymphocytes are therefore one of the most motile mammalian cell types, as they must be in “the right place at the right time”, making lymphocyte migration and function interdependent processes. Lymphoid organs are specialized tissues that provide the anatomic location and microenvironment in which lymphocytes achieve phenotypic and functional maturity (primary lymphoid organs) or undergo activation (secondary and tertiary lymphoid organs). Studies undertaken at the beginning of the 1990s showed that leukocyte extravasation from blood to tissues takes place in discrete steps, involving a sequence of receptor–ligand interactions, including selectin, integrins and chemokines. It was later observed that this model, defined as the multi-step paradigm, also applied for lymphocyte entry to SLO via high endothelial venules, a process also known as homing [1,4]. The activation signals that upregulate integrin adhesiveness on rolling leukocytes in a subsecond timescale are provided by chemokines. This family of small (8–12 kDa) polypeptides were originally described for their role in recruitment of innate immune cells and certain effector T cells during inflammatory processes [5]. The chemokines orchestrate physiological lymphocyte recirculation, as binding to their receptors on lymphocytes has two functional outcomes: first, certain chemokines induce integrin activation, which triggers intravascular adhesion of lymphocytes [6] and second, chemokine gradients induce directed cell migration. Chemokines thus coordinate immune responses by recruiting blood-borne cells into target tissues and by regulating
Eukaryotic cell migration is a highly coordinated process that requires the integration of extracellular signals for synchronized triggering of intracellular mechanisms that generate cell movement. Diverse physiological events, from embryonic development to wound healing, rely largely on efficient cell migration. The importance of cell migration and the contribution of various chemokines and receptors have been studied extensively, and the function of migration has been clearly established in the surveillance of the organism by cells of the immune system. For rapid and effective antigen recognition, each mature lymphocyte must sample the organism in a continuous search for its cognate antigen. To achieve this, naïve lymphocytes have evolved to migrate continuously, residing transiently in the so-called secondary lymphoid organs (SLO) strategically located throughout the body. From SLO, these cells return to the blood to continue their migration before returning to the SLO, a process known as lymphocyte recirculation [1–3]. Effector cells leave the SLO and migrate to non-lymphoid tissues, guided by local inflammatory signals elicited by pathogens [3].
∗ Corresponding author. Tel.: +34 915854852; fax: +34 913720493. E-mail address:
[email protected] (M. Mellado). 0165-2478/$ – see front matter © 2012 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.imlet.2012.04.012
24
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29
homeostasis through control of the microenvironmental organization of primary and SLO [3,5,7].
2. Chemokines: key molecules that govern cell movement The chemokines have been the focus of extraordinary interest over the last 25 years [8]. They are not only linked to lymphocyte trafficking, but also participate in regulation of T cell differentiation, HIV-1 infection, angiogenesis, tumorigenesis and development [9–13]. Nearly 50 chemokines have been described, which share remarkably similar three-dimensional structures despite their limited amino acid sequence similarity [9]. The original classification of chemokines based on structural criteria (C, CC, CXC and CX3C chemokines) is being abandoned, and replaced by a functional classification by which chemokines are grouped into two main categories, homeostatic or constitutive, and inflammatory or inducible [14]. In general, constitutive chemokines are regulated during development, whereas inducible chemokine expression is regulated mainly during inflammatory processes. In addition, several viruses encode highly selective chemokine receptor ligands that function as agonists or antagonists, and might thus have a role in viral dissemination or evasion of host immune responses [15]. Chemokines act by interacting with a group of seventransmembrane receptors coupled to G proteins of the GPCR family [8]. As for the chemokines, these receptors can also be grouped into two major families, CCR and CXCR, which interact with the CC and CXC chemokines, respectively [16]. There is apparently considerable promiscuity between chemokines and their receptors, with different ligands that can associate the same receptor and distinct receptors that can bind the same ligand. Furthermore, a single cell can express several chemokine receptors, simultaneously or during different stages of its life. Based on their broad range of functions, it is easy to deduce that chemokines must be central to a variety of diseases characterized by inflammation and cell infiltration [17] such as asthma, atherosclerosis, rheumatoid arthritis, multiple sclerosis, colitis, Crohn’s disease, experimental autoimmune encephalomyelitis (EAE) or psoriasis. Finally, CXCR4 and CCR5 are the two main co-receptors for HIV-1 infection [18], and some chemokine receptors also participate in tumor metastasis [12] and transplant rejection [19]. Given the correlation between the expression of specific chemokines and the specific recruitment of cell populations in the course of certain disease processes, the chemokines and their receptors have become a major focus in research and pharmaceutical laboratories. Their medical importance has led to intense effort to obtain structural information that could aid the development and optimization of therapeutics to inhibit their functions [20–22]. Conclusions drawn from animal models unfortunately cannot always be extrapolated to man, and although there are several molecules in clinical trials to target chemokines and their receptors in inflammatory diseases, results so far are very disappointing. Blockade of a chemokine or/and a chemokine receptor binding site might be an overly simplistic approach to altering chemokine function, and a reanalysis of chemokine biology is thus justified. Expression of chemokines and their receptors is finely regulated by several factors that include cytokines, growth factors, and cell cycle status [23–25], indicating that cell context influences these responses. The complexity of this scenario is completed by the glycosaminoglycans (GAG), which present chemokines to cells [26], and by the propensity of ligands and receptors to multimerize. Antagonists and neutralizing antibodies that are active in vitro are completely inactive when tested in vivo, possibly because they cannot recognize the structure of the chemokines or of their receptors in this context.
At nanomolar concentrations, some obligate chemokine monomers retain full activity for triggering receptor-mediated cell responses in vitro [27,28]. Several lines of evidence nonetheless indicate that at chemokine-rich inflammation sites in vivo, these proteins might also multimerize through GAG-mediated or -independent mechanisms, which would influence their cell activation properties [29]. As several excellent reviews have been published on the links between chemokine structure and function, these topics will not be reiterated here; instead, we limit our discussion to chemokine receptor conformation. 3. Chemokine receptors: a critical element for chemokine function 3.1. Introduction There are 20 signaling chemokine receptors, as well as three non-signaling scavenger receptors, all of which govern the immune response by binding, internalizing, and degrading chemokines [30,31], as well as several receptors encoded by viruses. All of these receptors are comprised of approximately 350 amino acids, with a molecular weight of approximately 40 kDa. The extracellular domain consists of the N terminus and three extracellular loops that act in concert to bind the chemokine ligand. The intracellular region is composed of three loops and the C terminus, which collaborate to transduce the chemokine signal. Based on their amino acid sequences, chemokine receptors belong to the class A rhodopsin-like family. Although similar to other seventransmembrane receptors, the chemokine receptors share certain structural features, such as the highly conserved DRYLAIV amino acid sequence in the second intracellular loop [32]; this motif is absent in the decoy receptors, indicating its implication in signaling. 3.2. Chemokine receptor oligomerization Several studies report that certain GPCR can function as monomers in vitro [33–35]; bioinformatic, structural and functional analyses have nonetheless advanced the concept that chemokine receptors are probably expressed at the cell surface as dimers/oligomers [36–40]. The crystal structure of CXCR4 in the presence of antagonists (a small molecule or a cyclic peptide) recently revealed a homodimeric conformation with the interface located in transmembrane regions V and VI [41]. Although initially considered a cytokine habit, evidence thus supports the existence of chemokine receptor homo- and heterodimers (Table 1). Chemokine receptor dimerization was first demonstrated for CCR2, the CCL2 receptor, using various experimental approaches that included agonist monoclonal antibodies, tagged receptors, and mutant receptors [42]. Coimmunoprecipitation and Western blot studies of chemokine receptors often detect individual receptors as well as high molecular weight species that could correspond to receptor complexes. These types of strategies nonetheless yield a static view of the system that can lead to misinterpretation of results. For example, in Western blot and coimmunoprecipitation, we detected CCR2 dimers only after ligand activation, leading us to conclude that ligands had an active role in promoting receptor dimerization [42], although later experiments using imaging-based techniques clearly showed that dimers form in the absence of chemokines [37,38,40]. The difficulty in detecting these complexes by immunoprecipitation might be due to instability of the dimer conformation in the absence of ligand, which cannot resist the detergent treatment. Analysis of live and fixed receptor-expressing cells using fluorescence and bioluminescence resonance energy transfer (FRET, BRET) techniques [43] greatly enhanced our understanding of
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29
25
Table 1 Chemokine receptor homo- and heterodimers. Experimental evidence supports chemokine receptor homo- and heterodimerization. Ab, antibody; BiFC, bimolecular fluorescence microscopy; BRET, bioluminescence resonance energy transfer; co-IP, co-immunoprecipitation; FLIM, fluorescence lifetime imaging microscopy; FRET, fluorescence resonance energy transfer; IS, immunological synapse; mAb, monoclonal antibody; PCA, protein fragment complementation assay; TM, transmembrane domain.
Chemokine receptor homodimerization
Receptor
Method
Cells
Comments
References
CCR2/CCR2
Co-IP
Cell line
First description of chemokine receptor dimerization
[42]
BRET BRET Co-IP Co-IP
Cell line Cell line Cell line Cell line
CCR5/CCR5
Cell line
CXCR1/CXCR1
CXCR2/CXCR2
FRET FLIM Co-IP Bio-informatic BiFC
Cell line and primary cells
Co-IP FRET Tr-FRET BRET FRET
Cell line
Co-IP
Co-IP FRET BRET FRET CXCR4/CXCR4
Co-IP BRET
Cell line
Cell line and primary cells Cell line and primary cells Cell line
Cell line and primary cells Primary cells Cell line
BRET Co-IP FRET
Cell line
BRET BRET-BiFC
Cell line
PCA
Cell line
BRET
Cell line
Crystalization CXCR7/CXCR7
Chemokine receptor heterodimerization
CCR2/CCR5
BRET
Cell line
PCA
Cell line
BRET
Cell Line
Co-IP FRET
Cell line
Co-IP
Cell line and primary cells
In silico (lipid-facing mutational analysis) Co-IP BRET BRET
CCR2/CXCR4
Co-IP FRET BRET
Cell line Cell line
Cell line Cell line
Anti-CCR5 Ab increase BRET Anti-CCR5 mAb induce dimerization CCR5 dimerization blocks HIV-1 infection CCR532 retains CCR5 in intracellular compartments Ligands stabilize preformed receptor dimer. TM1–TM4 peptides reduce FRET signals and block function
[38] [46] [81] [82] [51] [37]
[83] CCR5 homodimers signaling differs from that of CCR5/CXCR4 heterodimers by binding of a specific adaptor protein [40] CXCL8 does not promote changes in FRET signal
CXCL8 promotes conformational changes in homodimers GluR1 co-expression impairs dimer formation, CXCR2 deletion mutants act as dominant negative receptors CXCL8 does not promote changes in FRET signal
[45]
CXCL8 promotes conformational changes in homodimers
[45]
CXCR4 antagonists and CXCL12 alter BRET signal Ligand independent dimerization gp120 increases FRET, but AMD3100 decreases gp120-induced FRET in cells expressing hCD4 Higher-order oligomerization in a ligand independent fashion; TM implicated in oligomerization CXCL12 and pharmacological agents produced changes in bioluminescence in CXCR4/CXCR4 CXCL12 modifies the conformation of preformed CXCR4 homodimers First crystal structure for a chemokine receptor CXCL12 modifies the conformation of preformed CXCR7 homodimers Inhibitors and ligands enhance CXCR7 homodimerization AMD3100 is an allosteric agonist; CXCL12 induces conformational changes Ligands and mAb stabilize preformed receptor heterodimers, blocking HIV-1 entry Homo- and heterodimers activate distinct signaling pathways Bioinformatic analysis indicating the feasibility of CCR2/CCR5 heterodimers Cross-competition in ligand binding assays Chemokines do not influence the heterodimerization status. Negative binding cooperativity Anti-CCR2 mAb stabilizes preformed heterodimers, blocking HIV-1 entry CXCR4 TM peptides do not affect BRET signal. Ligand induces conformational changes in preformed dimers
[64]
[40]
[84] [38] [63] [85]
[36]
[86]
[76] [41] [76] [86] [87]
[88,89]
[72] [90] [56] [57]
[89] [38]
26
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29
Table 1 (Continued) Receptor
CCR5/CXCR4
CXCR1/CXCR2
CXCR4/CXCR7
CCR2/CCR5/CXCR4
Method
Cells
Comments
References
BRET
Co-IP
Cell line
Co-IP BRET
Cell line
FRET
Cell line
BRET
Cell line
BiFC
Cell line
Co-IP FRET BRET FRET BRET FRET BRET
Cell line
Allosteric transinhibition by specific inhibitors in the heterodimer Ligands induce receptor dimers; blocks HIV-1 entry CCR5 interferes with CXCR4 expression, endocytosis and HIV-1 co-receptor activity CCR5/CXCR4 heterodimers are recruited to the IS. Heterodimer is essential for the costimulatory effect in T cell activation Ligands promote conformational changes in the heterodimer Specific ligands promote conformational changes Heterodimer does not interact with NHERF1, hence CCR5/CXCR4 signaling differs from that of CCR5 homodimer Receptors form homo- as well as heterodimers
[58]
Co-IP
Cell line and primary cell Cell line
PCA BRET BiFC
Cell line and primary cells Cell line Cell line Cell line Cell line
receptor complexes at the cell surface. These methods are based on detection of receptor-coupled fluorophores excited by an external light source (FRET) or by enzyme catalysis of a luminescent substrate (BRET). Several groups used this technology to show that chemokine receptors form homodimers, complexes formed by the same receptor, and heterodimers, complexes formed by two distinct receptors. Photobleaching FRET experiments showed that all CXC chemokine receptors form homo- and heterodimers ˜ et al., submitted), and that FRET efficiency varies (Martínez-Munoz depending on the receptors analyzed. As this efficiency depends on the distance between and orientation of the fluorophores, these data indicate that the conformation adopted by a given receptor is ˜ et al., influenced by its coexpression with others (Martínez-Munoz submitted). FRET or BRET efficiency can also be determined at different ratios of donor:acceptor fluorophores coupled to receptors. These methods allow generation of saturated BRET or FRET curves that permit evaluation of the half-maximal BRET/FRET signal, that is, the relative affinity of receptor molecules in the complex. Apparent affinity calculations are used to study a specific receptor complex in a variety of experimental conditions, i.e., after ligand addition, coexpression of an unlabeled receptor, or cell/membrane modification [38,44,45]. We cannot compare the association force between different receptor complexes, however, as affinity difference is only one possible cause of the variations in FRET and BRET values [43]. As some of these methods allow evaluation of the subcellular locations of these complexes, we assume that homo- and heterodimers are found not only at the cell surface even in the absence of ligands, but also in intracellular vesicles, presumably formed during receptor synthesis and maturation [46]. As for other GPCR [47], CCR5 and CXCR4 complexes have been detected in small transGolgi vesicles [48], suggesting that receptor oligomerization is a mechanism that regulates receptor expression at the cell surface. This observation concurs with other GPCR studies suggesting that early dimerization in the endoplasmic reticulum (ER), which could have a role in trafficking, is a general feature of this receptor family [49]. CXCR2 coexpression with a mutant CXCR1 that is retained
CXCL8 promotes disruption of the heterodimer Synergic response to CXCL12 Heterodimer signals as independent entity, regulating CXCR4 signaling First evidence of chemokine receptor hetero-oligomerization
[88] [91]
[73]
[44] [54] [83]
[40]
[45] [78] [76] [86] [54]
in the ER causes a substantial reduction in cell surface expression [40]. It was hypothesized that, through dimerization with CCR5, the natural genetic mutant CCR532 retains the complex in the ER and prevents transport of normal CCR5 to the cell surface, retarding progression of HIV-1 infection [50,51]. Nonetheless, no data yet indicate whether dimerization facilitates chemokine receptor expression at the membrane, as for other GPCR [52]. Atomic force microscopy has been used to show that rhodopsin, the prototypical member of the class A GPCR, forms dimers that are condensed into supramolecular structures whose topography consists of densely packed lines [53]. Experiments using transfected cells indicate that chemokine receptor homo- and heterodimers form large receptor arrays on the cell membrane, although this remains to be validated in primary cells. At the cell membrane, CXCR4 engages in higher-order oligomerization in a ligand-independent fashion, as shown by a combination of BRET and bimolecular fluorescence complementation analysis [36]; this approach also demonstrated oligomers containing CXCR4, CCR5 and CCR2 [54]. The recent characterization of the CXCR4 crystal structure also supports interactions between CXCR4 dimers in higher order complexes [41]. Bioinformatic structure analysis of a CCR5 homology model based on the rhodopsin receptor allowed us to predict interaction of hydrophobic amino acid residues in the transmembrane regions [55]; studies of CXCR4 mutants lacking various domains [36], together with the crystal structure of CXCR4 bound to small antagonists, confirmed that chemokine receptor dimerization is promoted mainly by these interactions. Many questions remain to be answered. Do all chemokine receptors interact through similar regions, or are there dimer-specific dimerization regions depending on the receptors involved? Does homo- and heterodimerization of a given receptor always involve the same residues? Which regions are involved in the interactions between chemokine receptors in larger arrays? Is there any difference between receptor dimers alone or in the presence of ligand? The answers to these questions will be of great value in the design of new drugs able to modulate chemokine responses through interference with receptor dimerization.
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29
27
3.3. Chemokine receptor oligomers: dynamic structures One possible consequence of the existence of receptor arrays is allosteric modulation between chemokine receptor dimers. In transfected and primary cells coexpressing CCR2/CCR5, CCR5specific ligands prevent binding of CCR2-specific ligands, although they are unable to bind CCR2 coexpressed [56,57]; similar experiments show allosteric transmodulation for CCR2 and CXCR4 [58]. The CXCR4-specific antagonist AMD3100 blocks binding of CCR2specific ligands; conversely, the CCR2, CCR5, and CXCR3 antagonist TAK779 [59] strongly inhibits CXCL12 binding in cells that coexpress CXCR4 [58]. Results were similar when TAK779 and AMD3100 were tested in cells expressing multimeric CXCR4, CCR5 and CCR2 complexes [54]. An alternative explanation for allosteric modulation is that G protein might contact the partners of a dimeric complex in a non-symmetric manner [60], leading to a differential effect of G protein on agonist affinity for the receptors involved; this could result in negative cooperativity in agonist binding. Limitations in G protein pool availability might also explain negative ligand cooperativity; even without crosstalk between the receptors that form the complex, they could attempt to bind simultaneously to the same G protein [61]. These observations are difficult to interpret if we consider the chemokine receptors as individual entities. They nonetheless fit a model that we term the “cigar bundle” [62], in which multiple chemokine receptors are expressed at the cell surface in dynamic arrays; presence of a ligand affects not only its own specific receptor, but also other receptors in the same array. In accordance with this concept, we observed that CXCR1 and CXCR2 homo- and heterodimers are dynamically regulated both by receptor expression and by ligand binding [45]. Although FRET and BRET techniques are limited in their capacity to distinguish between conformational changes and complex disruption, FRET/BRET efficiency variations have been observed after ligand addition. CXCL12 addition increases BRET between CXCR4 homodimers [38], ligand binding to CXCR4 or CCR5 alters CXCR4/CCR5 heterodimers [44], and CXCL8 abolishes FRET signals for CXCR1/CXCR2 heterodimers while stabilizing values for the two homodimers [45]. Other laboratories nonetheless do not observe chemokine effects on receptor conformation [63]; for example, CCL2 does not promote conformational changes in CXCR4/CCR2 heterodimers [38].
4. Functional consequences of chemokine receptor dimerization Although we cannot rule out the possibility that chemokine receptor monomers can be operative, several indirect observations suggest that the dimer is the minimal functional unit of chemokine receptors. Synthetic peptides that prevent receptor dimerization reduce in vitro and in vivo function [37,39]. Some non-functional receptor mutants behave as dominant negative on wild type receptors; non-functional CCR2 and CXCR2 mutants block ligand-mediated cell migration by dimerizing with their wild type receptors [42,64]. Heterodimerization between CCR5 and the non-functional receptor DARC inhibits CCR5-mediated signaling [65]. Although there are no doubts about chemokine receptor oligomerization, evidence for the functional relevance of homoand heterodimerization is nonetheless far from conclusive (Fig. 1). The discovery that GPCR form heterodimers with distinct pharmacological properties raises fascinating possibilities regarding the pharmacological plasticity and diversity of these signaling systems. Receptor dimerization affects ligand binding affinity for some GPCR [66] and modulates signal transduction [67]; there are nonetheless few data indicating affinity change of a chemokine for a given receptor expressed alone or with other receptors. Changes in IC50 for
inactive dimers
active heterodimers couples to signaling molecules
active homodimer couples to signaling molecules
FUNCTIONAL CONSEQUENCES [40, 51] 2. Allosteric modulation
[56-59] [54] [72-74]
5. Modulation of ligand functions
[65,76-80]
Fig. 1. Functional relevance of chemokine receptor oligomerization. Chemokine receptors are organized into oligomeric structures termed arrays, which contain homo- and heterodimers, probably formed during receptor synthesis and maturation. Homo- and heterodimers are dynamic conformations regulated not only by receptor expression, but also by ligand levels. Ligand binding to the receptor complex stabilizes the active conformation and signaling is triggered; these conformational changes are also propagated to neighboring receptors. After the activated complex is internalized, the array relaxes and receptors assume the resting conformation. Although few reports analyze the functional relevance of chemokine receptor heterodimers, several studies suggest their involvement in regulation of receptor trafficking, modulation of ligand affinity, promotion of allosteric modulation, triggering of distinct signaling cascades and/or modulation of behavior of other receptors in the complex.
CXCL12 in cells expressing CXCR4 alone or with CCR5 (1.46 nM vs. 0.35 nM) suggest modulation of CXCL12 binding affinity for CXCR4 as a result of receptor heterodimerization. In contrast, in the same conditions, IC50 for CCL4 binding to CCR5 was unaffected by CXCR4 coexpression (2.19 nM vs. 2.21 nM) [54]. Although associated with cooperative downstream signaling pathways [68] rather than with effects on ligand binding affinity, synergism between CCR7 ligands and CXCL13 renders T cells more competent in response to migratory cues [69]. CCL5/CXCL4 complexes are involved in monocyte recruitment and retention at atheroma plaques [70], and specific in vivo and in vitro functions have been reported for the CXCL8–CXCL4, CCL22–CCL19 and CCL2–CCL8 chemokine pairs [69,71]. Recent results from several groups support the hypothesis of functional diversity between chemokine receptor homo- and heterodimers. CCR2/CCR5 heterodimers trigger specific signaling events such as G11 activation, which in turn promotes specific cell functions including increased cell adhesion [72]. CCR5/CXCR4 heterodimers are recruited to the immunological synapse, where they couple to Gq/11 proteins; this leads to T cell insensitivity to chemotactic gradients, enhanced proliferation and cytokine production, and formation of more stable conjugates [73,74]. An increasing number of reports assign a role to receptor oligomerization in chemokine function. Nonetheless, lack of specific tools for stabilizing heterodimeric complexes and the constantly changing equilibrium between receptor conformations complicate study of
28
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29
the signaling and function of each conformation, as well as its pharmacological implications. Several reports nonetheless indicate that receptor heterodimerization can modulate the behavior of individual partners. CXCR7 shares the CXCL12 ligand with CXCR4, but does not activate G␣i proteins [75,76]. CXCR7/CXCR4 heterodimers are reported in both transfectants and primary cells, where CXCR7 modulates CXCR4-mediated responses [76–78]; CXCR7/CXCR4 heterodimers should thus be considered distinct functional units with novel properties, which contribute to the functional plasticity of CXCL12. Similarly, DARC inhibits CCR5-mediated functions by forming DARC/CCR5 complexes [65]. Do these data suggest that the non-signaling receptors DARC, D6 or CXC–CKR can, in addition to their scavenging properties, modulate chemokine receptor function through dimerization? Functional interaction between chemokine receptors and other unrelated GPCR has also been described. Heterodimerization of CCR5 or CXCR4 with opioid receptors alters chemokine responses [79]. Pharmacological modulation of the CXCR2 receptor results in changes in DOR (delta opioid receptor) function; interestingly, a small molecule antagonist for CXCR2 seems to enhance signaling in response to DOR agonists, although the ligands do not compete directly for the other receptors [80]. 5. Conclusions Although much evidence supports a central role for chemokines in physiological and pathological processes and the pharmaceutical industry has made an extraordinary effort to develop chemokinebased therapeutic drugs [22], results so far are very disappointing. Blockade of a chemokine or/and a receptor binding site might be an overly simplistic approach to altering the function of many chemokines. Given the complex scenario of chemokine activity, with ligands and receptors forming complexes in dynamic equilibrium, with proteins that bind and modulate their functions, and with post-transductional modifications, one can speculate that antagonists designed in vitro might not recognize a specific motif in vivo. To better understand chemokine function and improve design of new therapeutic tools, it is time to consider factors that have not been taken into account previously. Work in recent years has provided evidence that chemokine receptors are found and might function as dimers. We must confirm the functional relevance and regulation of homo- and heterodimeric complexes in the cells, tissues, and organisms in which they are naturally expressed, since most current studies use heterologous expression systems. When these questions begin to be resolved, the formidable challenge of chemokine receptor oligomerization will offer new possibilities for the design of drug-screening and therapeutic strategies. Acknowledgements We thank people at the Chemokine signaling’s group for much of the work that contributed to this review, and to C. Bastos and C. Mark for secretarial and editorial assistance, respectively. This work was supported in part by grants from the Spanish Ministry of Science and Innovation (SAF 2008-03388), from Instituto de Salud Carlos III (ISCIII) program RETICS, RD08/0075 (RIER) (RD08/0075/0010) and the European Union (Innochem LSHB-CT-2005-518167 and FP7-integrated project Masterswitch no. 223404). References [1] Butcher EC, Picker LJ. Lymphocyte homing and homeostasis. Science 1996;272:60–6. [2] Gowans JL, Knight EJ. The Route of Re-Circulation of Lymphocytes in the Rat. Proc R Soc Lond B Biol Sci 1964;159:257–82.
[3] von Andrian UH, Mackay CR. T-cell function and migration. Two sides of the same coin. N Engl J Med 2000;343:1020–34. [4] Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 1994;76:301–14. [5] Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol 2004;22:891–928. [6] Campbell RN. Assessing the spread of engineered TMV. Science 1999;286:903. [7] Cyster JG. Chemokines and cell migration in secondary lymphoid organs. Science 1999;286:2098–102. [8] Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol 2000;18:217–42. [9] Clore GM, Gronenborn AM. Three-dimensional structures of alpha and beta chemokines. FASEB J 1995;9:57–62. [10] Fernandez EJ, Lolis E. Structure, function, and inhibition of chemokines. Annu Rev Pharmacol Toxicol 2002;42:469–99. [11] Mackay CR. Chemokines: immunology’s high impact factors. Nat Immunol 2001;2:95–101. [12] Muller 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. [13] Strieter RM. Chemokines: not just leukocyte chemoattractants in the promotion of cancer. Nat Immunol 2001;2:285–6. [14] Proudfoot AE. Chemokine receptors: multifaceted therapeutic targets. Nat Rev Immunol 2002;2:106–15. [15] Alcami A. Viral mimicry of cytokines, chemokines and their receptors. Nat Rev Immunol 2003;3:36–50. [16] Murphy PM, Baggiolini M, Charo IF, Hebert CA, Horuk R, Matsushima K, et al. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev 2000;52:145–76. [17] Murdoch C, Finn A. Chemokine receptors and their role in inflammation and infectious diseases. Blood 2000;95:3032–43. [18] Littman DR. Chemokine receptors: keys to AIDS pathogenesis. Cell 1998;93:677–80. [19] el-Sawy T, Fahmy NM, Fairchild RL. Chemokines: directing leukocyte infiltration into allografts. Curr Opin Immunol 2002;14:562–8. [20] Horuk R. Development and evaluation of pharmacological agents targeting chemokine receptors. Methods 2003;29:369–75. [21] Proudfoot AE, Power CA, Rommel C, Wells TN. Strategies for chemokine antagonists as therapeutics. Semin Immunol 2003;15:57–65. [22] Wells TN, Power CA, Shaw JP, Proudfoot AE. Chemokine blockers–therapeutics in the making. Trends Pharmacol Sci 2006;27:41–7. [23] Loetscher P, Seitz M, Baggiolini M, Moser B. Interleukin-2 regulates CC chemokine receptor expression and chemotactic responsiveness in T lymphocytes. J Exp Med 1996;184:569–77. [24] Papadopoulos EJ, Sassetti C, Saeki H, Yamada N, Kawamura T, Fitzhugh DJ, et al. Fractalkine, a CX3C chemokine, is expressed by dendritic cells and is up-regulated upon dendritic cell maturation. Eur J Immunol 1999;29:2551–9. [25] Parks E, Strieter RM, Lukacs NW, Gauldie J, Hitt M, Graham FL, et al. Transient gene transfer of IL-12 regulates chemokine expression and disease severity in experimental arthritis. J Immunol 1998;160:4615–9. [26] Johnson Z, Proudfoot AE, Handel TM. Interaction of chemokines and glycosaminoglycans: a new twist in the regulation of chemokine function with opportunities for therapeutic intervention. Cytokine Growth Factor Rev 2005;16:625–36. [27] Jin H, Shen X, Baggett BR, Kong X, LiWang PJ. The human CC chemokine MIP-1beta dimer is not competent to bind to the CCR5 receptor. J Biol Chem 2007;282:27976–83. [28] Rajarathnam K, Sykes BD, Kay CM, Dewald B, Geiser T, Baggiolini M, et al. Neutrophil activation by monomeric interleukin-8. Science 1994;264:90–2. [29] Baltus T, Weber KS, Johnson Z, Proudfoot AE, Weber C. Oligomerization of RANTES is required for CCR1-mediated arrest but not CCR5-mediated transmigration of leukocytes on inflamed endothelium. Blood 2003;102:1985–8. [30] Graham GJ, McKimmie CS. Chemokine scavenging by D6: a movable feast. Trends Immunol 2006;27:381–6. [31] Nibbs R, Graham G, Rot A. Chemokines on the move: control by the chemokine interceptors Duffy blood group antigen and D6. Semin Immunol 2003;15:287–94. [32] Murphy PM. The molecular biology of leukocyte chemoattractant receptors. Annu Rev Immunol 1994;12:593–633. [33] Bayburt TH, Vishnivetskiy SA, McLean MA, Morizumi T, Huang CC, Tesmer JJ, et al. Monomeric rhodopsin is sufficient for normal rhodopsin kinase (GRK1) phosphorylation and arrestin-1 binding. J Biol Chem 2011;286:1420–8. [34] Ernst OP, Gramse V, Kolbe M, Hofmann KP, Heck M. Monomeric G proteincoupled receptor rhodopsin in solution activates its G protein transducin at the diffusion limit. Proc Natl Acad Sci U S A 2007;104:10859–64. [35] Kuszak AJ, Pitchiaya S, Anand JP, Mosberg HI, Walter NG, Sunahara RK. Purification and functional reconstitution of monomeric mu-opioid receptors: allosteric modulation of agonist binding by Gi2. J Biol Chem 2009;284:26732–41. [36] Hamatake M, Aoki T, Futahashi Y, Urano E, Yamamoto N, Komano J. Ligand-independent higher-order multimerization of CXCR4, a G-proteincoupled chemokine receptor involved in targeted metastasis. Cancer Sci 2009;100:95–102. [37] Hernanz-Falcon P, Rodriguez-Frade JM, Serrano A, Juan D, del Sol A, Soriano SF, et al. Identification of amino acid residues crucial for chemokine receptor dimerization. Nat Immunol 2004;5:216–23.
L.M. Mu˜ noz et al. / Immunology Letters 145 (2012) 23–29 [38] Percherancier Y, Berchiche YA, Slight I, Volkmer-Engert R, Tamamura H, Fujii N, et al. Bioluminescence resonance energy transfer reveals ligand-induced conformational changes in CXCR4 homo- and heterodimers. J Biol Chem 2005;280:9895–903. [39] Wang J, He L, Combs CA, Roderiquez G, Norcross MA. Dimerization of CXCR4 in living malignant cells: control of cell migration by a synthetic peptide that reduces homologous CXCR4 interactions. Mol Cancer Ther 2006;5: 2474–83. [40] Wilson S, Wilkinson G, Milligan G. The CXCR1 and CXCR2 receptors form constitutive homo- and heterodimers selectively and with equal apparent affinities. J Biol Chem 2005;280:28663–74. [41] Wu B, Chien EY, Mol CD, Fenalti G, Liu W, Katritch V, et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science 2010;330:1066–71. [42] Rodriguez-Frade JM, Vila-Coro AJ, de Ana AM, Albar JP, Martinez AC, Mellado M. The chemokine monocyte chemoattractant protein-1 induces functional responses through dimerization of its receptor CCR2. Proc Natl Acad Sci U S A 1999;96:3628–33. [43] Kenworthy AK. Imaging protein-protein interactions using fluorescence resonance energy transfer microscopy. Methods 2001;24:289–96. [44] Isik N, Hereld D, Jin T. Fluorescence resonance energy transfer imaging reveals that chemokine-binding modulates heterodimers of CXCR4 and CCR5 receptors. PLoS One 2008;3:e3424. [45] Martinez Munoz L, Lucas P, Navarro G, Checa AI, Franco R, Martinez AC, et al. Dynamic regulation of CXCR1 and CXCR2 homo- and heterodimers. J Immunol 2009;183:7337–46. [46] Issafras H, Angers S, Bulenger S, Blanpain C, Parmentier M, Labbe-Jullie C, et al. Constitutive agonist-independent CCR5 oligomerization and antibodymediated clustering occurring at physiological levels of receptors. J Biol Chem 2002;277:34666–73. [47] Bulenger S, Marullo S, Bouvier M. Emerging role of homo- and heterodimerization in G-protein-coupled receptor biosynthesis and maturation. Trends Pharmacol Sci 2005;26:131–7. [48] Singer II, Scott S, Kawka DW, Chin J, Daugherty BL, DeMartino JA, et al. CCR5, CXCR4, and CD4 are clustered and closely apposed on microvilli of human macrophages and T cells. J Virol 2001;75:3779–90. [49] Angers S, Salahpour A, Bouvier M. Dimerization: an emerging concept for G protein-coupled receptor ontogeny and function. Annu Rev Pharmacol Toxicol 2002;42:409–35. [50] Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 1996;382:722–5. [51] Chelli M, Alizon M. Determinants of the trans-dominant negative effect of truncated forms of the CCR5 chemokine receptor. J Biol Chem 2001;276:46975–82. [52] White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, et al. Heterodimerization is required for the formation of a functional GABA(B) receptor. Nature 1998;396:679–82. [53] Fotiadis D, Liang Y, Filipek S, Saperstein DA, Engel A, Palczewski K. Atomicforce microscopy: rhodopsin dimers in native disc membranes. Nature 2003;421:127–8. [54] Sohy D, Yano H, de Nadai P, Urizar E, Guillabert A, Javitch JA, et al. Heterooligomerization of CCR2, CCR5, and CXCR4 and the protean effects of selective antagonists. J Biol Chem 2009;284:31270–9. [55] de Juan D, Mellado M, Rodriguez-Frade JM, Hernanz-Falcon P, Serrano A, del Sol A, et al. A framework for computational and experimental methods: identifying dimerization residues in CCR chemokine receptors. Bioinformatics 2005;21(Suppl. 2):ii13–8. [56] El-Asmar L, Springael JY, Ballet S, Andrieu EU, Vassart G, Parmentier M. Evidence for negative binding cooperativity within CCR5–CCR2b heterodimers. Mol Pharmacol 2005;67:460–9. [57] Springael JY, Le Minh PN, Urizar E, Costagliola S, Vassart G, Parmentier M. Allosteric modulation of binding properties between units of chemokine receptor homo- and hetero-oligomers. Mol Pharmacol 2006;69:1652–61. [58] Sohy D, Parmentier M, Springael JY. Allosteric transinhibition by specific antagonists in CCR2/CXCR4 heterodimers. J Biol Chem 2007;282:30062–9. [59] Gao P, Zhou XY, Yashiro-Ohtani Y, Yang YF, Sugimoto N, Ono S, et al. The unique target specificity of a nonpeptide chemokine receptor antagonist: selective blockade of two Th1 chemokine receptors CCR5 and CXCR3. J Leukoc Biol 2003;73:273–80. [60] Damian M, Martin A, Mesnier D, Pin JP, Baneres JL. Asymmetric conformational changes in a GPCR dimer controlled by G-proteins. EMBO J 2006;25: 5693–702. [61] Chabre M, Deterre P, Antonny B. The apparent cooperativity of some GPCRs does not necessarily imply dimerization. Trends Pharmacol Sci 2009;30:182–7. [62] Thelen M, Munoz LM, Rodriguez-Frade JM, Mellado M. Chemokine receptor oligomerization: functional considerations. Curr Opin Pharmacol 2010;10:38–43. [63] Babcock GJ, Farzan M, Sodroski J. Ligand-independent dimerization of CXCR4, a principal HIV-1 coreceptor. J Biol Chem 2003;278:3378–85. [64] Trettel F, Di Bartolomeo S, Lauro C, Catalano M, Ciotti MT, Limatola C. Ligandindependent CXCR2 dimerization. J Biol Chem 2003;278:40980–8.
29
[65] Chakera A, Seeber RM, John AE, Eidne KA, Greaves DR. The duffy antigen/receptor for chemokines exists in an oligomeric form in living cells and functionally antagonizes CCR5 signaling through hetero-oligomerization. Mol Pharmacol 2008;73:1362–70. [66] Jordan BA, Devi LA. G-protein-coupled receptor heterodimerization modulates receptor function. Nature 1999;399:697–700. [67] Terrillon S, Bouvier M. Roles of G-protein-coupled receptor dimerization. EMBO Rep 2004;5:30–4. [68] Gouwy M, Struyf S, Noppen S, Schutyser E, Springael JY, Parmentier M, et al. Synergy between coproduced CC and CXC chemokines in monocyte chemotaxis through receptor-mediated events. Mol Pharmacol 2008;74:485–95. [69] 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. [70] Koenen RR, von Hundelshausen P, Nesmelova IV, Zernecke A, Liehn EA, Sarabi A, et al. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice. Nat Med 2009;15:97–103. [71] Crown SE, Yu Y, Sweeney MD, Leary JA, Handel TM. Heterodimerization of CCR2 chemokines and regulation by glycosaminoglycan binding. J Biol Chem 2006;281:25438–46. [72] Mellado M, Rodriguez-Frade JM, Vila-Coro AJ, Fernandez S, Martin de Ana A, Jones DR, et al. Chemokine receptor homo- or heterodimerization activates distinct signaling pathways. EMBO J 2001;20:2497–507. [73] Contento RL, Molon B, Boularan C, Pozzan T, Manes S, Marullo S, et al. CXCR4–CCR5: a couple modulating T cell functions. Proc Natl Acad Sci U S A 2008;105:10101–6. [74] Molon B, Gri G, Bettella M, Gomez-Mouton C, Lanzavecchia A, Martinez AC, et al. T cell costimulation by chemokine receptors. Nat Immunol 2005;6:465–71. [75] Naumann U, Cameroni E, Pruenster M, Mahabaleshwar H, Raz E, Zerwes HG, et al. CXCR7 functions as a scavenger for CXCL12 and CXCL11. PLoS One 2010;5:e9175. [76] Levoye A, Balabanian K, Baleux F, Bachelerie F, Lagane B. CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling. Blood 2009;113:6085–93. [77] Sanchez-Alcaniz JA, Haege S, Mueller W, Pla R, Mackay F, Schulz S, et al. Cxcr7 controls neuronal migration by regulating chemokine responsiveness. Neuron 2011;69:77–90. [78] Sierro F, Biben C, Martinez-Munoz L, Mellado M, Ransohoff RM, Li M, et al. Disrupted cardiac development but normal hematopoiesis in mice deficient in the second CXCL12/SDF-1 receptor, CXCR7. Proc Natl Acad Sci U S A 2007;104:14759–64. [79] Pello OM, Martinez-Munoz L, Parrillas V, Serrano A, Rodriguez-Frade JM, Toro MJ, et al. Ligand stabilization of CXCR4/delta-opioid receptor heterodimers reveals a mechanism for immune response regulation. Eur J Immunol 2008;38:537–49. [80] Parenty G, Appelbe S, Milligan G. CXCR2 chemokine receptor antagonism enhances DOP opioid receptor function via allosteric regulation of the CXCR2DOP receptor heterodimer. Biochem J 2008;412:245–56. [81] Rodriguez-Frade JM, Vila-Coro AJ, Martin A, Nieto M, Sanchez-Madrid F, Proudfoot AE, et al. Similarities and differences in RANTES- and (AOP)RANTES-triggered signals: implications for chemotaxis. J Cell Biol 1999;144: 755–65. [82] Vila-Coro AJ, Mellado M, Martin de Ana A, Lucas P, del Real G, Martinez AC, et al. HIV-1 infection through the CCR5 receptor is blocked by receptor dimerization. Proc Natl Acad Sci U S A 2000;97:3388–93. [83] Hammad MM, Kuang YQ, Yan R, Allen H, Dupre DJ. Na+/H+ exchanger regulatory factor-1 is involved in chemokine receptor homodimer CCR5 internalization and signal transduction but does not affect CXCR4 homodimer or CXCR4–CCR5 heterodimer. J Biol Chem 2010;285:34653–64. [84] Vila-Coro AJ, Rodriguez-Frade JM, Martin De Ana A, Moreno-Ortiz MC, Martinez AC, Mellado M. The chemokine SDF-1alpha triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway. FASEB J 1999;13:1699–710. [85] Toth PT, Ren D, Miller RJ. Regulation of CXCR4 receptor dimerization by the chemokine SDF-1alpha and the HIV-1 coat protein gp120: a fluorescence resonance energy transfer (FRET) study. J Pharmacol Exp Ther 2004;310:8–17. [86] Luker KE, Gupta M, Luker GD. Imaging chemokine receptor dimerization with firefly luciferase complementation. FASEB J 2009;23:823–34. [87] Kalatskaya I, Berchiche YA, Gravel S, Limberg BJ, Rosenbaum JS, Heveker N. AMD3100 is a CXCR7 ligand with allosteric agonist properties. Mol Pharmacol 2009;75:1240–7. [88] Mellado M, Rodriguez-Frade JM, Vila-Coro AJ, de Ana AM, Martinez AC. Chemokine control of HIV-1 infection. Nature 1999;400:723–4. [89] Rodriguez-Frade JM, del Real G, Serrano A, Hernanz-Falcon P, Soriano SF, VilaCoro AJ, et al. Blocking HIV-1 infection via CCR5 and CXCR4 receptors by acting in trans on the CCR2 chemokine receptor. EMBO J 2004;23:66–76. [90] Gouldson PR, Dean MK, Snell CR, Bywater RP, Gkoutos G, Reynolds CA. Lipidfacing correlated mutations and dimerization in G-protein coupled receptors. Protein Eng 2001;14:759–67. [91] Wang J, Alvarez R, Roderiquez G, Guan E, Norcross MA. Constitutive association of cell surface CCR5 and CXCR4 in the presence of CD4. J Cell Biochem 2004;93:753–60.