Skip to main content

The chemokine receptor CCR5: multi-faceted hook for HIV-1

Abstract

Chemokines are cytokines whose primary role is cellular activation and stimulation of leukocyte migration. They perform their various functions by interacting with G protein-coupled cell surface receptors (GPCRs) and are involved in the regulation of many biological processes such as apoptosis, proliferation, angiogenesis, hematopoiesis or organogenesis. They contribute to the maintenance of the homeostasis of lymphocytes and coordinate the function of the immune system. However, chemokines and their receptors are sometimes hijacked by some pathogens to infect the host organism. For a given chemokine receptor, there is a wide structural, organizational and conformational diversity. In this review, we describe the evidence for structural variety reported for the chemokine receptor CCR5, how this variability can be exploited by HIV-1 to infect its target cells and what therapeutic solutions are currently being developed to overcome this problem.

Introduction

Chemokines belong to the cytokine family. Their chemoattractant properties are at the origin of their name (chemoattractant cytokines). They are a family of small, soluble 8–14 KDa proteins that exert important functions of intercellular communication. All chemokines exert their functions by binding to G-protein coupled receptors (GPCR) also called chemokine receptors. Chemokines, regulate many biological processes such as proliferation, apoptosis, angiogenesis, hematopoiesis or lymphoid organ development, but their main role is to activate and control leukocyte migration. Following interaction with their specific chemokine ligands, chemokine receptors trigger signaling that initiates a chemotaxis process, which traffics the cell to a desired location within the organism. These molecules are involved in the maintenance of lymphocyte homeostasis under physiological conditions and the deregulation of their expression is associated to many diseases [1]. Although chemokine receptors play a pivotal role in the host's antimicrobial defense mechanisms, they can serve as primary targets for various pathogens. These pathogens may either encode chemokine mimics or, as observed in the context of HIV-1, use chemokine receptors to directly infect their target cells.

Some pathogens can induce the production of molecules that bind to receptors with a higher affinity than their natural ligands [2,3,4,5], others will modify the expression level of chemokines [6, 7] or of chemokine receptors [8]. In all cases, this is an effective way to either evade immune selection or, on the opposite, to lead to a deleterious burst of the immune system (cytokine storm) [9]. This strategy is used by a wide variety of pathogens ranging from ticks [10, 11], parasites [12, 13] to bacteria [6, 7, 14] and viruses [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30]. Among viruses, one can cite as examples Epstein-Barr viruses that encode a GPCR (BILF1) capable of forming a dimer with CXCR4 chemokine receptor thereby inhibiting its signaling but also human cytomegalovirus (CMV) that secretes a soluble chemokine receptor (pUL21.5) which binds selectively to the chemokine RANTES with very high affinity, blocking the interaction of RANTES with its cellular receptors (CCR5). In both cases, viruses interfere with cellular signaling processes, reducing the effectiveness of the immune response [17]. Such phenomenon has also been described for poxviruses such as Respiratory syncytial virus [18], mousepox [19, 20], smallpox [21, 22], monkeypox [23] (of which several hundred cases have been diagnosed in humans in Europe during the spring 2022), herpes viruses [24,25,26,27] and HIV [28]. This is not an exhaustive list and for more details see [29, 30].

In some cases, direct attachment of pathogens to a chemokine receptor represents the first step of infection, the receptor acting as an anchor that allows the pathogen to dock to the plasma membrane of its target cell before entering into the cell or releasing its contents into the cytoplasm. This has for instance been described for pathogenic bacteria Staphylococcus aureus (whose toxins interacts with CCR5 chemokine receptor) [31], for Human Respiratory Syncytial Virus (that binds to CX3CR1 chemokine receptor) [32,33,34] and HIV (that uses CCR5 and/or CXCR4 chemokine receptors to attach and infect immune cells) [35,36,37,38]. In the case of HIV-1, the CCR5 chemokine receptor not only acts as an anchor for the virus to attach to the cell surface, but also plays an active role in the infection process by initiating signaling cascades that mimics the signaling of chemokines and promotes infection [28, 39,40,41]. To be thorough, and although it is outside the scope of this review, it is interesting to note that the HIV can also bind to other receptors such as glycan-binding proteins, so-called lectins, which mediate potent viral transmission to CD4+ T cells known as trans-infection [42]. One can cite DC-SIGN/CD209 [43], the C-type lectin DC immunoreceptor (DCIR) [44], the mannose receptor (MR) or Siglec-1/CD169 [45].

A very large number of studies have been conducted on HIV-1 and have revealed the ability of this virus to exploit the wide structural variability (i.e. different conformational states) of chemokine receptors to bypass the immune system and circumvent some therapeutic approaches. It is this last aspect that we aim to discuss in this review: how the structural diversity of chemokine receptors is exploited by HIV-1 to infect its target cells, whether this diversity comes from one person to another, from one cell type to another or within the same cell. We will here focus mainly on the C-C chemokine receptor type 5, also known as CCR5 and to a lesser extent on C-X-C chemokine receptor type 4 (CXCR4).

HIV-1 infection requires interaction with chemokine receptors

The early steps of HIV-1 infection first involve the binding of the HIV protein envelope gp120 to the CD4 receptor (Fig. 1, step 1). This process then requires the interaction of gp120 with a chemokine receptor than can be either CCR5 or CXCR4 (Fig. 1, step 2). This second interaction defines the tropism of the virus infecting the cell; viruses that bind to CCR5 are called “R5”, those that bind to CXCR4 are called “X4” and those that are able to bind to both are known as “R5X4”. R5 viruses are preferentially transmitted [46] and are predominant during the chronic phase of infection [47, 48]. Approximately 50% of patients with advanced HIV develop X4 virus in addition to R5 virus. This usually occurs after several years and is associated with a decline in the CD4 + T cell count and a rapid progression to a pathologic state [49,50,51,52,53].

Fig. 1
figure 1

Schematic view of the early steps of HIV infection. The HIV envelope (Env), consisting of gp120 and gp41 subunits, first binds to the CD4 receptor located at the plasma membrane of the infected cell (1). This binding induces a conformational change in gp120 that allows the interaction of its V3 loop with a co-receptor (2). The co-receptor can be either CCR5 or CXCR4, defining the tropism of the virus (see main text). This second binding induces a new conformational change in gp120 which exposes the fusion peptide of gp41 that anchors its N-terminal domain into the plasma membrane of the target cell (3). This initiates a process of fusion that will lead to the release of the viral content into the cytoplasm of the infected cell (for more details see [38, 54])

The combination of the structural diversity of both HIV-1 and CCR5 offers a broad range of possible interactions between these molecules, providing as many opportunities for infection. On the one hand, the V3 loop of the viral protein gp120, which is involved in the binding to CCR5 (Fig. 1, step 2) [55, 56], is one of the most variable sites of HIV-1, with differences of up to 50% between isolates [57, 58]. This explains why the number of CCR5 molecules recognized by different HIV-1 gp120 proteins varies differently between various cell types [59]. On the other hand, GPCRs have long been depicted as switches that can exist in two distinct states: inactive and active. Over the past two decades, this vision has considerably evolved and it is now well established that the situation is far more complex than originally thought. Numerous crystallographic and cryo-electron microscopy structures have revealed that GPCRs can adopt many different three-dimensional structures. Ligands or G-proteins selectively bind to a subset of these conformations, causing a population shift and establishing a new equilibrium [60,61,62,63,64]. In this way, Urvas and Kellenberger recently published a comparative analysis of all chemokine—chemokine receptor structures uncovering that while N termini receptors structures exhibit conserved interaction patterns with chemokines, the second extracellular loop exhibits several subfamily-specific features [65]. Moreover, these structures are only the tip of the iceberg since they represent only static snapshots of the most stable states of the proteins when there is a thermal equilibrium which leads to oscillations between multiple transient conformations [66].

Structural variability of chemokines receptors involved in HIV-1 infection

CCR5 and CXCR4 receptors belong to the GPCR class A family. These cell-surface receptors can detect extracellular molecules and induce an intracellular response. Binding of signal molecules to the extracellular surface of the receptor activates intracellular G proteins which in turn act on various effectors mediating intracellular signaling effects. These receptors are made up of 7 transmembrane helices connected by three extracellular and three intracellular loops and their overall 3D structure can be modulated by many factors. Importantly, the 3D structure of CCR5 and CXCR4 associated with different antagonists has been elucidated a decade ago [67, 68].

Post-translational modifications

A first level of structural differences relates to the modulation of post-translational modifications of receptors. Hundreds of post-translational modifications of proteins have been reported to date [69]. This diversity leads to a structural variety of proteins that will influence their ability to interact with their ligands but also offer many possibilities for viruses to adapt to their attachment. Many post-translational modifications have been described for CCR5 including, o-glycosilation, phosphorylation, sulfation and palmitoylation (Fig. 2A).

Fig. 2
figure 2

Schematic representation of the factors that can influence the 3D-structure of CCR5 coreceptor. A Post-translational modifications (PMTs): sites of O-Glycosylation (Ser6, Thr16, Ser17) [70, 71], sulfation (Tyr3, 10, 14, 15) [73, 75,76,77,78], palmitoylation (Cys321, 323 and 324) [79,80,81,82,83,84,85,86,87,88,89,90,91] and phosphorylation (Ser336, 337, 342 and 349) [235, 236] are represented by colored circles. Factors such as G-protein coupling (B) [28, 60, 97,98,99,100,101,102,103], mutations (C) [126,127,128,129, 177, 178], surface density [117,118,119,120,121,122,123] and oligomerization [136,137,138,139,140,141,142,143,144,145,146,147,148,149] (D) and membrane surrounding [152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173] (E) are also exemplified

O-glycosylation and Phosphorylation—O-glycosylation of CCR5 does not appear to affect the ability of HIV-1 to infect cells [70] and there is little data on the impact of CCR5 phosphorylation on HIV-1 infection. Phosphorylation of this receptor appears to be primarily involved in internalization and recycling and thus its abundance at the surface of the cell (for review, see [71]). However, one cannot exclude that it also has an impact on the overall structure of the proteins and contributes to their structural diversity at the surface of the cell.

Sulfation—By contrast, sulfation of tyrosine residues 3, 10, 14 and 15 of the N-terminal moiety of CCR5 has been described to play an important role in the functionality of this receptor [72]. In 1999, Farzan and colleagues have shown that sulfation of the amino-terminal part of CCR5 contributes to the binding of its ligands MIP-1 α and MIP-1 β but also to its binding to gp120/CD4 complexes and thus to the ability of HIV-1 to infect cells [73]. More recently, using ELISA approaches and performing binding experiments on cells expressing CCR5 under conditions that modulate their sulfation level, Scurci and colleagues have shown that CCR5 sulfation is heterogeneous thereby affecting the binding properties of natural chemokines and some monoclonal antibodies (mAb 3A9 only binds to the sulfated peptide while mAb HEK/1/85a shows a clear preference for the unsulfated peptide) [74]. These authors also suggest that the difference in binding ability between natural ligands (MIP-1 α, MIP-1 β, RANTES/CCL5) and the analogue 5P12-RANTES (an HIV entry inhibitor that binds to CCR5) are—at least in part—due to the heterogeneity of sulfation on the cell surface (other factors, such as the molecular fit of ligands with the transmembrane domain of the receptor, can also influence their global affinity). This proposal is in agreement with other recent NMR works that have identified sulfation combinations that are important for ligand binding [75,76,77]. To a lesser extent this effect is also observed with CXCR4, which exists in several different forms of sulfation [78].

Palmitoylation—CCR5 can also be palmitoylated on three cysteine residues: C321, C323, and C324, in the C-terminal tail of CCR5 [79, 80]. The presence of palmitate groups will affect CCR5 at three levels: on the structure of the receptor itself, on its cell distribution and on its membrane localization [81, 82]. From a strictly structural perspective, the insertion of palmitate into the cytoplasmic leaflet of the plasma membrane profoundly modifies the conformation of the cytosolic tail housing the palmitoylated cysteine [83] and affect the overall GPCR structure [84,85,86]. In addition, such modification has been shown to promote coupling to G-receptors [87] and phosphorylation of CCR5 [88] leading to a potential increase of structural diversity of the receptors. Another important effect of palmitoylation lies in its impact on the membrane distribution of receptors within the cell. Palmitoylation of membrane proteins is often required for their correct folding, which protects them from the degradation pathway associated with the endoplasmic reticulum. In other words, palmitoylation orchestrates the subcellular traffic of GPCRs to the plasma membrane. For this reason, HIV-1 cannot infect cells in which CCR5 is not palmitoylated, because it is not addressed to the plasma membrane [79, 89]. The presence of a saturated lipid (i.e. palmitate) that inserts into the membrane is known to promote protein relocation into liquid-ordered raft domains [86, 90, 91]. In this regard, palmitoylation of CCR5 is essential for the incorporation of CCR5 into the plasma membrane rafts [85] that offers a new lipid environment to the proteins inducing some conformational changes that will be discussed in the following section of this manuscript.

Membrane organization of chemokine receptors

As mentioned before, chemokine receptors present a structural diversity linked to their intrinsic chemical composition. Beyond this strictly “structural” aspect, the receptor surrounding plays an essential role in the modulation of its three-dimensional structure. Among these environmental factors, the coupling with G proteins or other transmembrane proteins but also the interactions with their lipid environment are key elements.

Coupling to G proteins—As indicated by their name, GPCRs can be coupled (or not) with different types of heterotrimeric G proteins (Fig. 2B). Human genome encodes 18 different Gα proteins, 5 Gβ proteins, and 12 Gγ proteins [92]. CCR5 has been shown to interact with several of them: Gαi, Gαq, Gβ and Gγ [28, 93]. This combination of possible interactions not only contributes to the structural diversity of CCR5, offering larger possibilities of HIV to bind to the cells, but it also favors infection by triggering signal transduction upon HIV binding.

Conformational changes: many authors have studied if interactions with G proteins could modify CCR5 conformation and affect their ability to interact with HIV-1. Certain small molecule inhibitors of CCR5 used as HIV-1 inhibitors, such as vicriviroc (VVC), maraviroc (MVC) or TAK779, act as allosteric modulators that stabilize a conformation of CCR5 that the virus does not effectively recognize, thus preventing its entry [94,95,96]. Several authors have investigated whether the binding of these inhibitors was affected by the coupling of CCR5 with G proteins. By performing binding assays, they showed that the association of the intracellular domains of CCR5 with G proteins alters their interactions with the inhibitors, suggesting that the association of CCR5 with the signaling machinery induces substantial conformational changes that affect the binding of these inhibitors and their ability to block HIV-1 entry [97,98,99,100]. Similar conclusion were also obtained by other studies that used monoclonal antibodies able to identify different CCR5 conformations [101]. These proposals were corroborated by cryo-electron microscopy structures which showed notable structural changes depending on whether CCR5 was coupled to a Gαi1 protein or not [102]. This is also in agreement with former results obtained on another GPCR receptor, the β2-adrenergic receptor, whose coupling with the G protein is loose and does not stabilize the receptor in a single conformation but allows for the existence of distinct intermediates [60, 103]. Taken together, these results show that whether or not CCR5 is coupled to G proteins is a source of conformational diversity.

Signal transduction: binding of HIV-1 gp120 to CCR5 has been shown to trigger activation of Gαi or Gαq proteins. This results in the modulation of many important cellular functions, including cell migration, adhesion, or survival [104,105,106,107,108,109,110,111,112]. Additionally, it is noteworthy that the interaction between the viral envelope and CCR5 can also trigger a response via β-arrestins. Initially considered for their role in receptor desensitization, β-arrestins are now recognized as versatile scaffolds interacting with various signaling proteins, including MAPKs, PI3K, and protein kinase B [113]. The activated signaling pathways will ultimately impact the cell susceptibility to HIV infection. For instance, it has been demonstrated that exposure to viral gp120 can initiate viral replication in cultures of resting CD4 T cells from infected individuals [114]. These findings suggest that virus-hijacked signaling process may subsequently facilitate pathogenesis. More details of these processes can be found in reviews by Wu and Yoder [28], Nickolof-Bybel et al. [40] and Juno and Fowke [115].

The intricate interplay between the G proteins, the CCR5 receptor and its binding to the HIV envelope protein underscores the complexity of understanding the exact role of each in HIV infection. The ability of the virus to infect its target cells depends not only on its ability to bind to the receptor, but also on its ability to trigger a cellular response that promotes its spread. Therefore, elucidating not only the ability of the virus to bind to CCR5, but also the signaling pathways it induces in conjunction with these structural changes could open the door to the discovery of innovative antiretroviral agents.

Receptors surface density—HIV-1 infection involves lateral organization of the receptors as it requires the binding of trimers of the virus envelope glycoprotein to host cell receptors leading to membrane fusion. Numerous studies have shown that effective infection can only occur if several of these trimers are simultaneously involved in the process [116]. As a consequence, the density of receptors present at the surface of the cells will have a direct impact on the capacity of the viruses to infect them [117,118,119,120,121] (Fig. 2D). For example, by quantifying CCR5 expression in acutely HIV-infected subjects over a 2 year period, it was shown by Yang et al. that high levels of CCR5 on CD4 central memory cells are associated with rapid disease progression [122]. Conversely, in elite controllers (individuals who spontaneously control HIV-1 replication in the absence of antiretroviral therapy), the analysis of the surface phenotype and transcriptional profile of CD4 + T cells has revealed low CCR5 expression, leading to the conclusion that low CCR5 expression protects the cells from HIV-1 infection [123]. Finally, it has been shown that some mutations induce a decrease for CCR5 addressed to the plasma membrane, conferring to individuals an almost total resistance to HIV-1 infection. This is the case for truncated CCR5 due to the delta 24 mutation [124, 125] and the delta 32 mutation [126,127,128,129] (Fig. 2C) that are known to impact the localization of CCR5. This will be developed in the next chapter. Various studies have then investigated the minimum number of receptors required for infection. Infectivity assays performed on HeLa cells in which individual clones express either large or small amounts of CD4 and distinct amounts of CCR5 have shown that the minimum amount of CCR5 allowing infection is dependent on the quantity of CD4 in the membrane and vary from 1.103 to 1.104 CCR5 per cell [119]. Furthermore, approximately four to six CCR5s assemble around the virus to form a complex needed for infection [130]. Since the efficiency of infection depends on both CD4 and CCR5 concentrations rather than on precise amounts of each, the authors have proposed that this efficiency also depends on the diffusion of these proteins within the membrane and on their ability to encounter each other to form virus-CD4-CCR5 ternary complexes [37]. This was later on evidenced by Baker et al. who measured the dynamics of CD4 and CCR5 by FRAP and revealed the existence of membrane domains that concentrate CD4 and CCR5 in a 1:5 ratio [131]. Finally yet importantly, it appears that the levels and expression profile of CCR5 can influence the tropism of virus strains for different cell types [132,133,134] and correlates with the appearance of viral reservoirs [135].

Oligomerization of receptors—Many GPCRs are able to interact with each other to form higher-order structures (homomers and/or heteromers) (Fig. 2D) that can, upon allosteric modulation, exhibit distinct biochemical properties from monomers [136,137,138,139]. Both CCR5 and CXCR4 have been shown to form many different homo and heteromers whose exhaustive lists can be found in [140] (Table 1) and [141] (Table 1). Many studies have used Bioluminescence or Fluorescence Energy Transfer (BRET and FRET, respectively) to detect oligomers formation. These methods are based on the fact that light energy can only be transferred from a donor to an acceptor if these molecules are less than 10 nm apart. By using these experimental approaches, it has first been shown that CCR5 constitutively forms homodimers at the surface of T-lymphocytes [142]. Thereafter, similar fluorescence measurements combined with the use of monoclonal antibodies that recognize different epitopes of CCR5 allowed to establish that CCR5 oligomers are structurally different from monomers and that HIV-1 preferentially recognize the monomeric form of the receptor [143]. Similar results have been obtained by dynamic approaches consisting in tracking and classifying the motion of different receptor subpopulations [144, 145]. Further studies finally provided evidence of three distinct CCR5 dimeric organizations [146]. On the same principle, the formation of CCR5-CCR2b heteromers has been evidenced and binding experiments revealed that the interaction between heterodimer units is of allosteric nature and modifies the binding properties of chemokines [147, 148]. Interestingly, oligomerization of CCR5/CD4/CXCR4 receptors has also been reported in the literature. This association has been shown to prevent X4 HIV-1 virus from binding to target cells [149]. Overall, these data support the view that oligomerization of chemokine receptors affects their structure and may influence the susceptibility of cells to infection. The development of super-resolution fluorescence methods, such as Stochastic Optical Reconstruction Microscopy (STORM) or Photo-Activation Localization Microscopy (PALM) has permitted direct visualization of GPCRs such as mu-opioid receptor [150] and dopamine receptor [151] at the single-molecule level, unveiling that their oligomers are transitory structures with rapid association and dissociation kinetics. This opens the gate to future experiment carried out on CCR5 and CXCR4 to decipher their mechanism of oligomerization and its role in HIV-1 infection.

Lipid surrounding—Finally, it has long been known that the lipid membrane has an impact on the structuring of membrane proteins [152, 153] (Fig. 2E), this is particularly true for GPCRs [154,155,156], including CCR5 and CXCR4 [157,158,159,160]. For example, it has recently been reported that ceramide or related sphingolipids might invert the topology of CCR5, preventing macrophages from migrating toward CCL5 [161] and similar results were obtained with CXCR4 and the CXCL12 chemokine [160]. Concurrently, Calmet et al. have reconstituted CCR5 receptors into model membranes of controlled lipid composition and demonstrated that cholesterol decreases the binding affinity of maraviroc to CCR5 [157]. Molecular dynamics simulation further suggested that cholesterol restricts the structural dynamics of the receptor [157]. It is interesting to notice that sphingolipids and cholesterol are key components involved in the formation of lipid rafts [162, 163]. Actually, a lot of work has been done to understand the contribution of lipid rafts in the organization of HIV-1 receptors and in the infection process [164]. Indeed, extensive studies conducted in the late 1990s have shown that the CD4 receptor is clearly associated with rafts [165,166,167]. Subsequently, other studies based on raft extraction and protein analysis have shown that CCR5 is mainly associated with lipid rafts [168] while CXCR4 is only partially localized into them [168,169,170]. Moreover, the use of conformational antibodies has revealed that rafts stabilize the active conformation of CCR5 towards its ligands [171]. Lastly, Yang and colleagues have used model systems mimicking HIV-1 envelopes and T-cell membranes to demonstrate that the hydrophobic mismatch at boundaries between liquid-ordered and liquid-disordered regions (i.e. lipid raft and non-raft regions) generates a line tension that can facilitate membrane fusion with the virus envelope [172, 173]. Taken together, these data suggest that lipid rafts may act as platforms that facilitate virus entry both by fostering local concentrations of receptors and/or co-receptors and by modulating their conformation.

Genetic mutation of chemokine receptors is double-edged sword for HIV-1

In this review, we have discussed the beneficial effects that heterogeneity in chemokine receptor structure could have on HIV-1 infection. Nevertheless, this variability can, in some rare cases, be detrimental for viruses. Indeed, it has been found in the 90’s that about 1% of the Caucasian human population carries a homozygous 32 base pair deletion in the gene encoding for the CCR5 receptor, resulting in a truncated gene that produces non-functional receptors named CCR5 delta-32 [127, 132, 174]. This mutation, discovered in a group of homozygous patients known as "exposed uninfected", confers resistance to infection. It has been shown that the protein CCR5 delta-32 remains mostly localized in the endoplasmic reticulum where it exerts a trans-dominant negative effect on the wild type CCR5 protein, preventing its transport to the cell surface [175]. Furthermore, it forms dimers with the CXCR4 proteins in the ER, which also limits their distribution to the plasma membrane [176]. As a consequence, both CCR5 and CXCR4 are absent from the plasma membrane conferring to the cells a resistance to R5, X4 and R5X4 viruses [126,127,128,129]. This observation has been exploited to initially cure two patients who became famous and known as " Berlin patient" [177] and " London patient " [178]. Both of them have received allogeneic hematopoietic stem cell transplantation from donors with a homozygous CCR5 delta-32 mutation to cure a leukemia [179]. More recently, three other patients have been cured using the same strategy, one at Weill Cornell hospital of New York in the frame of IMPAACT P1107 clinical trial (NCT02140944) [180], a second one at the City of Hope cancer institute in Los Angeles as revealed by Jana Dickter during the AIDS 2022 press conference and a third one in Düsseldorf in 2023 [181]. At this stage, it should be stressed that we cannot exclude the possibility that additional factors, such as the transplant procedure itself, may have played a role in these results.

Although large-scale therapy based on this strategy is not feasible, these results demonstrate that CCR5 is a suitable target for HIV-1 gene therapy. It should be noted that such a treatment could potentially influence other medical conditions. For instance, it has been suggested that the CCR5-delta32 mutation might contribute to prolonged kidney transplant survival [182]. Conversely, it has also been proposed that this mutation could increase susceptibility to neuroinvasive West Nile virus infections [183].

Targeting CCR5 to fight the virus

In the last decades, three main strategies targeting CCR5 have mainly been developed to block HIV infection: the use of CCR5 ligands, the use of antibodies and, more recently, editing the genome in order to eradicate CCR5 from the cell surface or modify its structure. It is important to highlight that the first two strategies can take advantage of the structural diversity of CCR5 by specifically targeting the subset of receptors implicated in HIV-1 infection. This targeted approach may help to minimize the adverse effects of drugs. In the light of the structural variabilities of CCR5 discussed above, we will now discuss the potential of each of these strategies.

Development of CCR5 ligands—This strategy aims to develop molecules that will bind to CCR5 and prevent the interaction between CCR5 and the gp120 HIV protein thereby aborting HIV entry mediated by fusion and infection. Up today, many molecules have been produced such as Tak-779 [96], cenicriviroc [184], CMPD-167 [185], Aplaviroc [186], vicriviroc [94] or maraviroc [95] (for a detailed review see [187, 188]). Despite the large number of molecules developed and clinical trials carried out, only maraviroc has been approved for use in HIV treatment in 2007 [189]. It remains the only CCR5 inhibitor in clinical use to date. It is an inverse CCR5 agonist that impairs HIV envelope binding to CCR5 through a noncompetitive and allosteric modulation [67, 95, 98, 190].

However, nine CCR5 mutations have been identified as inducing a decrease of the affinity for maraviroc (up to tenfold) [191]. It should be noted that, with one exception, all these mutations also modify gp120 binding and are therefore less detrimental in terms of protection against viral infection. The use of maraviroc can cause a tropism switch to X4 strains although this is a very rare event in the absence of pre-existing X4 strains. This drug cannot be used in the presence of X4 strains, as a result, it requires a viral tropism test before use. Part of the problem of its use also arises from the possible emergence of virus variants that recognize the maraviroc-CCR5 complex, as well as free CCR5 [192,193,194].

Development of anti-CCR5 antibodies—Various attempts have been made to develop antibodies that prevent HIV infection [195,196,197]. These antibodies are designed not only to bind to CCR5 and prevent its interaction with the viral protein gp120, but also to trigger the internalization of the co-receptor [198]. Only two antibodies reached clinical trials: CCR5mAb004 [188], that have been withdrawn in 2006, and Leronlimab [196] (formerly PRO 140), for which many clinical trials have been completed (NCT02175680, NCT00110591, NCT02483078, NCT00613379, NCT00642707) or are still running (NCT03902522, NCT02859961, NCT02355184, NCT02990858, NCT05271370). This antibody has not yet been granted approval by the Food and Drug Administration, but has been designated fast-track status in 2019 [199]. Leronimab is a humanized monoclonal IgG4 antibody that targets the extracellular domain 2 of CCR5, and impairs its interaction with HIV-1 gp120 preventing virus entry. Subcutaneous administration of this antibody to HIV-1-infected individuals significantly reduced their viral load between successive injections [200]. When administered intravenously to HIV-1 infected adult subjects, it has also showed potent and long-lived antiviral activity [201]. The only downside is that Leronimab does not recognize all CCR5 proteins since receptor occupancy was around 85% [201] meaning that 15% of CCR5 receptors potentially escape to the antibody and might participate to the emergence of resistant HIV-1 strains. Their use also raises pharmacokinetic issues. Because they are proteins, they cannot be absorbed from the gastrointestinal tract and must be administered parenterally. A final issue arises from the fact that the use of anti-CCR5 antibodies can lead to a shift in tropism towards CXCR4 [202].

CCR5 gene editing—Recent advances in genome editing offers unprecedented possibilities for gene therapy, as it is now possible to replace one or more bases in any gene of interest, at any location [203, 204]. The CRISPR/Cas9 system or the use of zinc finger nucleases (ZFN) are methods of choice for such gene editing [205, 206]. These tools can be very useful to fight against HIV infection. The experimental strategy consists in extracting hematopoietic stem cells from patients, suppressing CCR5 expression in these cells thanks to an appropriate treatment (i.e. CRISPR/Cas9 or ZFN) before reinjecting them. The treated cells, newly resistant to infection, should then be able to proliferate and substitute cells sensitive to HIV-1. Several attempts have been performed in the frame of clinical trials such as NCT03164135 (CCR5 depletion thanks to CRIPR/CAS9), NCT03666871 and NCT02388594 (CCR5 depletion thanks to ZFN). These experiments have been shown to produce in vitro HIV-1 resistance which constitutes an encouraging proof of concept for HIV cure [207,208,209,210,211]. Despite these successes for CCR5 ablation, low genome-editing frequencies [212] and high off-target activity[213] have been observed. Therefore, genome-editing techniques still need to be improved before they can be used on a mass scale. Furthermore, since CCR5 deletion alone has no impact on infection by X4-tropic viruses, some authors have performed a double knockout (KO) CCR5 + CXCR4 [214]. Unfortunately, a poor engraftment of the R5X4-CRISPRCas9 KO CD4 + T cells in mice bone marrow has been obtained.

Another promising approach consists in taking advantage of the delta-32 mutation described earlier. This time, the strategy consists in modifying CCR5 gene (and not suppress it) in order to express the delta-32 truncated CCR5 protein. We have seen that this mutation suppresses the expression of CCR5 and CXCR4 at the cell surface, thus protecting them from R5- and X4-tropic viruses. This also present the advantage to target one gene instead of two and the risk of adverse side-effects seems limited insofar as this mutation has been observed naturally on thousands of people who do not seem to present any deleterious effects. Using this method, Ye et al. [215] and Qi et al. [216] succeeded to render cells resistant to HIV-1 infection whatever the tropism of the virus. Again unfortunately, the successful rate was low (20% on Jurkat cells and 11% in primary CD4 + cells) [216] so the protocols still need to be improved. Unlike the other approaches discussed in this chapter, CCR5 delta 32 editing overcomes the structural variability of CCR5 described in this manuscript. As such, this strategy appears to be a very promising approach for eradicating HIV by targeting CCR5.

Conclusion

The more varied the receptor structure, the more difficult it is to block them all, and the more opportunities viruses will have to bind to them to infect cells [123, 217]. The factors that can modulate the conformation and the organization of receptors involved in HIV-1 infection are numerous: post-translational modifications, expression level, coupling to G proteins, oligomerization or mutation of CCR5 and lipid environment (Fig. 2). These factors can be interdependent which further complicates their possible structural combinations (for instance, palmitoylation promotes the localization of CCR5 into the lipid rafts domains, which will then promote their interaction with the G proteins that are also concentrated there). All these parameters can vary within the same cell [98, 218,219,220,221,222,223], from one cell type to another [99, 224,225,226] and, by extent, from one patient to another. The combination of these factors leads to an infinite number of intermediate conformations which will present different affinities for their natural substrates but also for HIV-1 receptors [67, 99, 100, 221, 227,228,229,230], themselves presenting a very large structural variety [59, 231,232,233]. This tremendous diversity explains the difficulties encountered over the past 30 years in the fight against AIDS. Considerable advances have been made and HIV infection is now well controlled by antiretroviral therapy (ART) and patients can live almost normally as long as they have access to these treatments and adhere strictly to the medication instructions. Nevertheless, these treatments do not block viral entry and they are not without risk since they can induce numerous side effects such as central nervous system disorders, bone defects, cardiovascular, hepatic or renal risks [234]. In addition, the treatments options remain limited for some patients with multiclass resistance and must be taken for life as they do not target the integrated proviral genome and fail to eradicate the virus in so called viral reservoirs. From this point of view, recent advances in genome editing offer encouraging prospects not only for blocking viral entry by modifying CCR5, but also for excising HIV sequences integrated into the genome [204]. One can hope that the combination of all these new therapeutic approaches will bring us closer to the development of an effective antiviral treatment against HIV that will be affordable for the broadest possible populations.

Availability of data and materials

Not applicable.

References

  1. Raman D, Sobolik-Delmaire T, Richmond A. Chemokines in health and disease. Exp Cell Res. 2011;317:575–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Drayman N, Glick Y, Ben-nun-Shaul O, Zer H, Zlotnick A, Gerber D, et al. Pathogens use structural mimicry of native host ligands as a mechanism for host receptor engagement. Cell Host Microbe. 2013;14:63–73.

    Article  CAS  PubMed  Google Scholar 

  3. Lüttichau HR, Stine J, Boesen TP, Johnsen AH, Chantry D, Gerstoft J, et al. A highly selective CC chemokine receptor (CCR)8 antagonist encoded by the poxvirus molluscum contagiosum. J Exp Med. 2000;191:171–80.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Pontejo SM, Murphy PM. Chemokines encoded by herpesviruses. J Leukoc Biol. 2017;102:1199–217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chensue SW. Molecular machinations: chemokine signals in host-pathogen interactions. Clin Microbiol Rev. 2001;14:821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sprenger H, Krause A, Kaufmann A, Priem S, Fabian D, Burmester GR, et al. Borrelia burgdorferi induces chemokines in human monocytes. Infect Immun. 1997;65:4384–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Thompson D, Sorenson J, Greenmyer J, Brissette CA, Watt JA. The Lyme disease bacterium, Borrelia burgdorferi, stimulates an inflammatory response in human choroid plexus epithelial cells. PLoS ONE. 2020;15:e0234993.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Secchiero P, Zella D, Capitani S, Gallo RC, Zauli G. Extracellular HIV-1 tat protein up-regulates the expression of surface CXC-chemokine receptor 4 in resting CD4+ T cells. J Immunol. 1999;162:2427–31.

    Article  CAS  PubMed  Google Scholar 

  9. Atasheva S, Shayakhmetov DM. Cytokine responses to adenovirus and adenovirus vectors. Viruses. 2022;14:888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Déruaz M, Frauenschuh A, Alessandri AL, Dias JM, Coelho FM, Russo RC, et al. Ticks produce highly selective chemokine binding proteins with antiinflammatory activity. J Exp Med. 2008;205:2019–31.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Denisov SS, Ippel JH, Heinzmann ACA, Koenen RR, Ortega-Gomez A, Soehnlein O, et al. Tick saliva protein evasin-3 modulates chemotaxis by disrupting CXCL8 interactions with glycosaminoglycans and CXCR2. J Biol Chem. 2019;294:12370–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Smith P, Fallon RE, Mangan NE, Walsh CM, Saraiva M, Sayers JR, et al. Schistosoma mansoni secretes a chemokine binding protein with antiinflammatory activity. J Exp Med. 2005;202:1319–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ofir-Birin Y, Ben Ami Pilo H, Cruz Camacho A, Rudik A, Rivkin A, Revach O-Y, et al. Malaria parasites both repress host CXCL10 and use it as a cue for growth acceleration. Nat Commun. 2021;12:4851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lambey P, Otun O, Cong X, Hoh F, Brunel L, Verdié P, et al. Structural insights into recognition of chemokine receptors by Staphylococcus aureus leukotoxins. Elife. 2022;11:e72555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Guo X, Feng Y, Zhao X, Qiao S, Ma Z, Li Z, et al. Coronavirus porcine epidemic diarrhea virus utilizes chemokine interleukin-8 to facilitate viral replication by regulating Ca2+ flux. J Virol. 2023;97:e0029223.

    Article  PubMed  Google Scholar 

  16. Koyanagi N, Kawaguchi Y. Evasion of the cell-mediated immune response by alphaherpesviruses. Viruses. 2020;12:1354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rosenkilde MM, Tsutsumi N, Knerr JM, Kildedal DF, Garcia KC. Viral G protein-coupled receptors encoded by β- and γ-herpesviruses. Annu Rev Virol. 2022;9:329–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Johnson SM, McNally BA, Ioannidis I, Flano E, Teng MN, Oomens AG, et al. Respiratory syncytial virus uses CX3CR1 as a receptor on primary human airway epithelial cultures. PLoS Pathog. 2015;11:e1005318.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Heidarieh H, Alcamí A. Mechanism of action of the viral chemokine-binding protein E163 from ectromelia virus. J Biol Chem. 2018;293:17418–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Alcami A, Saraiva M. Chemokine binding proteins encoded by pathogens. Adv Exp Med Biol. 2009;666:167–79.

    Article  CAS  PubMed  Google Scholar 

  21. Dunlop LR, Oehlberg KA, Reid JJ, Avci D, Rosengard AM. Variola virus immune evasion proteins. Microbes Infect. 2003;5:1049–56.

    Article  CAS  PubMed  Google Scholar 

  22. Carfí A, Smith CA, Smolak PJ, McGrew J, Wiley DC. Structure of a soluble secreted chemokine inhibitor vCCI (p35) from cowpox virus. Proc Natl Acad Sci USA. 1999;96:12379–83.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Hernáez B, Alonso-Lobo JM, Montanuy I, Fischer C, Sauer S, Sigal L, et al. A virus-encoded type I interferon decoy receptor enables evasion of host immunity through cell-surface binding. Nat Commun. 2018;9:5440.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Nijmeijer S, Leurs R, Smit MJ, Vischer HF. The Epstein-Barr virus-encoded G protein-coupled receptor BILF1 hetero-oligomerizes with human CXCR4, scavenges Gαi proteins, and constitutively impairs CXCR4 functioning. J Biol Chem. 2010;285:29632–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang D, Bresnahan W, Shenk T. Human cytomegalovirus encodes a highly specific RANTES decoy receptor. Proc Natl Acad Sci U S A. 2004;101:16642–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gershengorn MC, Geras-Raaka E, Varma A, Clark-Lewis I. Chemokines activate Kaposi’s sarcoma-associated herpesvirus G protein-coupled receptor in mammalian cells in culture. J Clin Invest. 1998;102:1469–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Vischer HF, Nijmeijer S, Smit MJ, Leurs R. Viral hijacking of human receptors through heterodimerization. Biochem Biophys Res Commun. 2008;377:93–7.

    Article  CAS  PubMed  Google Scholar 

  28. Wu Y, Yoder A. Chemokine coreceptor signaling in HIV-1 infection and pathogenesis. PLoS Pathog. 2009;5:e1000520.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Heidarieh H, Hernáez B, Alcamí A. Immune modulation by virus-encoded secreted chemokine binding proteins. Virus Res. 2015;209:67–75.

    Article  CAS  PubMed  Google Scholar 

  30. Pontejo SM, Murphy PM, Pease JE. Chemokine subversion by human herpesviruses. J Innate Immun. 2018;10:465–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Alonzo F, Kozhaya L, Rawlings SA, Reyes-Robles T, DuMont AL, Myszka DG, et al. CCR5 is a receptor for Staphylococcus aureus leukotoxin ED. Nature. 2013;493:51–5.

    Article  PubMed  Google Scholar 

  32. Green G, Johnson SM, Costello H, Brakel K, Harder O, Oomens AG, et al. CX3CR1 is a receptor for human respiratory syncytial virus in cotton rats. J Virol. 2021;95:e0001021.

    Article  PubMed  Google Scholar 

  33. Zhivaki D, Lemoine S, Lim A, Morva A, Vidalain P-O, Schandene L, et al. Respiratory syncytial virus infects regulatory b cells in human neonates via chemokine receptor CX3CR1 and promotes lung disease severity. Immunity. 2017;46:301–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Feng Z, Xu L, Xie Z. Receptors for respiratory syncytial virus infection and host factors regulating the life cycle of respiratory syncytial virus. Front Cell Infect Microbiol. 2022;12:858629.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dragic T, Litwin V, Allaway G, Martin S, Huang Y, Nagashima K, et al. HIV-1 entry into CD4 sup + cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381:667–73.

    Article  CAS  PubMed  Google Scholar 

  36. Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature. 1996;381:661–6.

    Article  CAS  PubMed  Google Scholar 

  37. Dumas F, Preira P, Salomé L. Membrane organization of virus and target cell plays a role in HIV entry. Biochimie. 2014;107(Pt A):22–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dumas F, Haanappel E. Lipids in infectious diseases—the case of AIDS and tuberculosis. Biochim Et Biophys Acta (BBA) Biomemb. 2017;1859:1636–47.

    Article  CAS  Google Scholar 

  39. François F, Klotman ME. Phosphatidylinositol 3-kinase regulates human immunodeficiency virus type 1 replication following viral entry in primary CD4+ T lymphocytes and macrophages. J Virol. 2003;77:2539–49.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology. 2021. https://doi.org/10.1186/s12977-021-00569-x.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Lucera MB, Fleissner Z, Tabler CO, Schlatzer DM, Troyer Z, Tilton JC. HIV signaling through CD4 and CCR5 activates Rho family GTPases that are required for optimal infection of primary CD4+ T cells. Retrovirology. 2017;14:4.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Dufloo J, Bruel T, Schwartz O. HIV-1 cell-to-cell transmission and broadly neutralizing antibodies. Retrovirology. 2018;15:51.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Tsegaye TS, Pöhlmann S. The multiple facets of HIV attachment to dendritic cell lectins. Cell Microbiol. 2010;12:1553–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lambert AA, Barabé F, Gilbert C, Tremblay MJ. DCIR-mediated enhancement of HIV-1 infection requires the ITIM-associated signal transduction pathway. Blood. 2011;117:6589–99.

    Article  CAS  PubMed  Google Scholar 

  45. Izquierdo-Useros N, Lorizate M, McLaren PJ, Telenti A, Kräusslich H-G, Martinez-Picado J. HIV-1 capture and transmission by dendritic cells: the role of viral glycolipids and the cellular receptor Siglec-1. PLoS Pathog. 2014;10:e1004146.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Shaw GM, Hunter E. HIV transmission. Cold Spring Harb Perspect Med. 2012;2:a006965.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Keele BF, Giorgi EE, Salazar-Gonzalez JF, Decker JM, Pham KT, Salazar MG, et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc Natl Acad Sci USA. 2008;105:7552–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Joseph SB, Swanstrom R. The evolution of HIV-1 entry phenotypes as a guide to changing target cells. J Leukoc Biol. 2018;103:421–31.

    Article  CAS  PubMed  Google Scholar 

  49. Koot M, Keet IP, Vos AH, de Goede RE, Roos MT, Coutinho RA, et al. Prognostic value of HIV-1 syncytium-inducing phenotype for rate of CD4+ cell depletion and progression to AIDS. Ann Intern Med. 1993;118:681–8.

    Article  CAS  PubMed  Google Scholar 

  50. Connor RI, Sheridan KE, Ceradini D, Choe S, Landau NR. Change in coreceptor use correlates with disease progression in HIV-1–infected individuals. J Exp Med. 1997;185:621–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Connell BJ, Hermans LE, Wensing AMJ, Schellens I, Schipper PJ, van Ham PM, et al. Immune activation correlates with and predicts CXCR4 co-receptor tropism switch in HIV-1 infection. Sci Rep. 2020;10:15866.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Moyle GJ, Wildfire A, Mandalia S, Mayer H, Goodrich J, Whitcomb J, et al. Epidemiology and predictive factors for chemokine receptor use in HIV-1 infection. J Infect Dis. 2005;191:866–72.

    Article  PubMed  Google Scholar 

  53. Weinberger AD, Perelson AS. Persistence and emergence of X4 virus in HIV infection. Math Biosci Eng. 2011;8:605–26.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Wilen CB, Tilton JC, Doms RW. HIV: cell binding and entry. Cold Spring Harb Perspect Med. 2012;2:a006866.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hwang SS, Boyle TJ, Lyerly HK, Cullen BR. Identification of the envelope V3 loop as the primary determinant of cell tropism in HIV-1. Science. 1991;253:71–4.

    Article  CAS  PubMed  Google Scholar 

  56. Garcia-Perez J, Staropoli I, Azoulay S, Heinrich J-T, Cascajero A, Colin P, et al. A single-residue change in the HIV-1 V3 loop associated with maraviroc resistance impairs CCR5 binding affinity while increasing replicative capacity. Retrovirology. 2015;12:50.

    Article  PubMed  PubMed Central  Google Scholar 

  57. LaRosa GJ, Davide JP, Weinhold K, Waterbury JA, Profy AT, Lewis JA, et al. Conserved sequence and structural elements in the HIV-1 principal neutralizing determinant. Science. 1990;249:932–5.

    Article  CAS  PubMed  Google Scholar 

  58. Wahlberg J, Albert J, Lundeberg J, Von Gegerfelt A, Broliden K, Utter G, et al. Analysis of the V3 loop in neutralization-resistant human immunodeficiency virus type 1 variants by direct solid-phase DNA sequencing. AIDS Res Hum Retrovir. 1991;7:983–90.

    Article  CAS  PubMed  Google Scholar 

  59. Colin P, Zhou Z, Staropoli I, Garcia-Perez J, Gasser R, Armani-Tourret M, et al. CCR5 structural plasticity shapes HIV-1 phenotypic properties. PLoS Pathog. 2018;14:e1007432.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Weis WI, Kobilka BK. The molecular basis of G protein-coupled receptor activation. Annu Rev Biochem. 2018;87:897–919.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ring AM, Manglik A, Kruse AC, Enos MD, Weis WI, Garcia KC, et al. Adrenaline-activated structure of β2-adrenoceptor stabilized by an engineered nanobody. Nature. 2013;502:575–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Rasmussen SGF, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, et al. Crystal structure of the β2 adrenergic receptor–Gs protein complex. Nature. 2011;477:549–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Koehl A, Hu H, Maeda S, Zhang Y, Qu Q, Paggi JM, et al. Structure of the µ-opioid receptor–Gi protein complex. Nature. 2018;558:547–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Cherezov V, Rosenbaum DM, Hanson MA, Rasmussen SGF, Thian FS, Kobilka TS, et al. High-resolution crystal structure of an engineered human β2-adrenergic G protein-coupled receptor. Science. 2007;318:1258–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Urvas L, Kellenberger E. Structural insights into molecular recognition and receptor activation in chemokine-chemokine receptor complexes. J Med Chem. 2023;66:7070–85.

    Article  CAS  PubMed  Google Scholar 

  66. Casiraghi M. Biophysical investigations of class A GPCRs. Biochimie. 2022;S0300–9084(22):00256–65.

    Google Scholar 

  67. Tan Q, Zhu Y, Li J, Chen Z, Han GW, Kufareva I, et al. Structure of the CCR5 chemokine receptor—HIV entry inhibitor maraviroc complex. Science. 2013. https://doi.org/10.1126/science.1241475.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Wu B, Chien EYT, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Garavelli JS. The RESID database of protein modifications as a resource and annotation tool. Proteomics. 2004;4:1527–33.

    Article  CAS  PubMed  Google Scholar 

  70. Bannert N, Craig S, Farzan M, Sogah D, Santo NV, Choe H, et al. Sialylated O-glycans and sulfated tyrosines in the NH2-terminal domain of CC chemokine receptor 5 contribute to high affinity binding of chemokines. J Exp Med. 2001;194:1661–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Patwardhan A, Cheng N, Trejo J. Post-translational modifications of G protein-coupled receptors control cellular signaling dynamics in space and time. Pharmacol Rev. 2021;73:120–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cormier EG, Persuh M, Thompson DA, Lin SW, Sakmar TP, Olson WC, et al. Specific interaction of CCR5 amino-terminal domain peptides containing sulfotyrosines with HIV-1 envelope glycoprotein gp120. Proc Natl Acad Sci USA. 2000;97:5762–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Farzan M, Mirzabekov T, Kolchinsky P, Wyatt R, Cayabyab M, Gerard NP, et al. Tyrosine sulfation of the amino terminus of CCR5 facilitates HIV-1 entry. Cell. 1999;96:667–76.

    Article  CAS  PubMed  Google Scholar 

  74. Scurci I, Akondi KB, Pinheiro I, Paolini-Bertrand M, Borgeat A, Cerini F, et al. CCR5 tyrosine sulfation heterogeneity generates cell surface receptor subpopulations with different ligand binding properties. Biochim Biophys Acta Gen Subj. 2021;1865:129753.

    Article  CAS  PubMed  Google Scholar 

  75. Abayev M, Rodrigues JPGLM, Srivastava G, Arshava B, Jaremko Ł, Jaremko M, et al. The solution structure of monomeric CCL5 in complex with a doubly sulfated N-terminal segment of CCR5. FEBS J. 2018;285:1988–2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kessler N, Akabayov SR, Moseri A, Cohen LS, Sakhapov D, Bolton D, et al. Allovalency observed by transferred NOE: interactions of sulfated tyrosine residues in the N-terminal segment of CCR5 with the CCL5 chemokine. FEBS J. 2021;288:1648–63.

    Article  CAS  PubMed  Google Scholar 

  77. Huang C-C, Lam SN, Acharya P, Tang M, Xiang S-H, Hussan SS-U, et al. Structures of the CCR5 N terminus and of a tyrosine-sulfated antibody with HIV-1 gp120 and CD4. Science. 2007;317:1930–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Farzan M, Babcock GJ, Vasilieva N, Wright PL, Kiprilov E, Mirzabekov T, et al. The role of post-translational modifications of the CXCR4 amino terminus in stromal-derived factor 1α association and HIV-1 entry *. J Biol Chem. 2002;277:29484–9.

    Article  CAS  PubMed  Google Scholar 

  79. Blanpain C, Wittamer V, Vanderwinden JM, Boom A, Renneboog B, Lee B, et al. Palmitoylation of CCR5 is critical for receptor trafficking and efficient activation of intracellular signaling pathways. J Biol Chem. 2001;276:23795–804.

    Article  CAS  PubMed  Google Scholar 

  80. Percherancier Y, Planchenault T, Valenzuela-Fernandez A, Virelizier JL, Arenzana-Seisdedos F, Bachelerie F. Palmitoylation-dependent control of degradation, life span, and membrane expression of the CCR5 receptor. J Biol Chem. 2001;276:31936–44.

    Article  CAS  PubMed  Google Scholar 

  81. Jansen M, Beaumelle B. How palmitoylation affects trafficking and signaling of membrane receptors. Biol Cell. 2022;114:61–72.

    Article  CAS  PubMed  Google Scholar 

  82. Zhang B, Li S, Shui W. Post-translational modifications of G protein-coupled receptors revealed by proteomics and structural biology. Front Chem. 2022;10:843502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Chini B, Parenti M. G-protein-coupled receptors, cholesterol and palmitoylation: facts about fats. J Mol Endocrinol. 2009;42:371–9.

    Article  CAS  PubMed  Google Scholar 

  84. Joseph M, Nagaraj R. Interaction of peptides corresponding to fatty acylation sites in proteins with model membranes. J Biol Chem. 1995;270:16749–55.

    Article  CAS  PubMed  Google Scholar 

  85. Chalhoub G, McCormick PJ. Palmitoylation and G-protein coupled receptors. Prog Mol Biol Transl Sci. 2022;193:195–211.

    Article  CAS  PubMed  Google Scholar 

  86. Charollais J, Van Der Goot FG. Palmitoylation of membrane proteins (Review). Mol Membr Biol. 2009;26:55–66.

    Article  CAS  PubMed  Google Scholar 

  87. Wedegaertner PB, Chu DH, Wilson PT, Levis MJ, Bourne HR. Palmitoylation is required for signaling functions and membrane attachment of Gq alpha and Gs alpha. J Biol Chem. 1993;268:25001–8.

    Article  CAS  PubMed  Google Scholar 

  88. Kraft K, Olbrich H, Majoul I, Mack M, Proudfoot A, Oppermann M. Characterization of sequence determinants within the carboxyl-terminal domain of chemokine receptor CCR5 that regulate signaling and receptor internalization. J Biol Chem. 2001;276:34408–18.

    Article  CAS  PubMed  Google Scholar 

  89. Boncompain G, Herit F, Tessier S, Lescure A, Del Nery E, Gestraud P, et al. Targeting CCR5 trafficking to inhibit HIV-1 infection. Sci Adv. 2019;5:0821.

    Article  Google Scholar 

  90. Levental I, Lingwood D, Grzybek M, Coskun U, Simons K. Palmitoylation regulates raft affinity for the majority of integral raft proteins. Proc Natl Acad Sci USA. 2010;107:22050–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Blaskovic S, Blanc M, van der Goot FG. What does S-palmitoylation do to membrane proteins? FEBS J. 2013;280:2766–74.

    Article  CAS  PubMed  Google Scholar 

  92. Syrovatkina V, Alegre KO, Dey R, Huang X-Y. Regulation, signaling and physiological functions of G-proteins. J Mol Biol. 2016;428:3850–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lorenzen E, Ceraudo E, Berchiche YA, Rico CA, Fürstenberg A, Sakmar TP, et al. G protein subtype-specific signaling bias in a series of CCR5 chemokine analogs. Sci Signal. 2018;11:6152.

    Article  Google Scholar 

  94. Strizki JM, Tremblay C, Xu S, Wojcik L, Wagner N, Gonsiorek W, et al. Discovery and characterization of vicriviroc (SCH 417690), a CCR5 antagonist with potent activity against human immunodeficiency virus type 1. Antimicrob Agents Chemother. 2005;49:4911–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Dorr P, Westby M, Dobbs S, Griffin P, Irvine B, Macartney M, et al. Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob Agents Chemother. 2005;49:4721–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Baba M, Nishimura O, Kanzaki N, Okamoto M, Sawada H, Iizawa Y, et al. A small-molecule, nonpeptide CCR5 antagonist with highly potent and selective anti-HIV-1 activity. Proc Natl Acad Sci USA. 1999;96:5698–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Berro R, Yasmeen A, Abrol R, Trzaskowski B, Abi-Habib S, Grunbeck A, et al. Use of G-protein-coupled and -uncoupled CCR5 receptors by CCR5 inhibitor-resistant and -sensitive human immunodeficiency virus type 1 variants. J Virol. 2013;87:6569–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Garcia-Perez J, Rueda P, Staropoli I, Kellenberger E, Alcami J, Arenzana-Seisdedos F, et al. New insights into the mechanisms whereby low molecular weight CCR5 ligands inhibit HIV-1 infection. J Biol Chem. 2011;286:4978–90.

    Article  CAS  PubMed  Google Scholar 

  99. Berro R, Klasse PJ, Lascano D, Flegler A, Nagashima KA, Sanders RW, et al. Multiple CCR5 conformations on the cell surface are used differentially by human immunodeficiency viruses resistant or sensitive to CCR5 inhibitors. J Virol. 2011;85:8227–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Colin P, Bénureau Y, Staropoli I, Wang Y, Gonzalez N, Alcami J, et al. HIV-1 exploits CCR5 conformational heterogeneity to escape inhibition by chemokines. Proc Natl Acad Sci USA. 2013;110:9475–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Flegler AJ, Cianci GC, Hope TJ. CCR5 conformations are dynamic and modulated by localization, trafficking and G protein association. PLoS ONE. 2014;9:e89056.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Zhang H, Chen K, Tan Q, Shao Q, Han S, Zhang C, et al. Structural basis for chemokine recognition and receptor activation of chemokine receptor CCR5. Nat Commun. 2021;12:4151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Yao XJ, Vélez Ruiz G, Whorton MR, Rasmussen SGF, DeVree BT, Deupi X, et al. The effect of ligand efficacy on the formation and stability of a GPCR-G protein complex. Proc Natl Acad Sci USA. 2009;106:9501–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Balabanian K, Harriague J, Décrion C, Lagane B, Shorte S, Baleux F, et al. CXCR4-tropic HIV-1 envelope glycoprotein functions as a viral chemokine in unstimulated primary CD4+ T lymphocytes. J Immunol. 2004;173:7150–60.

    Article  CAS  PubMed  Google Scholar 

  105. Yoder A, Yu D, Dong L, Iyer SR, Xu X, Kelly J, et al. HIV envelope-CXCR4 signaling activates cofilin to overcome cortical actin restriction in resting CD4 T cells. Cell. 2008;134:782–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Smith LK, Babcock IW, Minamide LS, Shaw AE, Bamburg JR, Kuhn TB. Direct interaction of HIV gp120 with neuronal CXCR4 and CCR5 receptors induces cofilin-actin rod pathology via a cellular prion protein- and NOX-dependent mechanism. PLoS ONE. 2021;16:e0248309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Gao R, Fang Q, Zhang X, Xu Q, Ye H, Guo W, et al. R5 HIV-1 gp120 activates p38 MAPK to induce rat cardiomyocyte injury by the CCR5 coreceptor. Pathobiology. 2019;86:274–84.

    Article  CAS  PubMed  Google Scholar 

  108. Wu Y. Flow cytometry-based quantification of HIV-induced T cell chemotactic response. Methods Mol Biol. 2016;1407:207–15.

    Article  CAS  PubMed  Google Scholar 

  109. Del Cornò M, Donninelli G, Varano B, Da Sacco L, Masotti A, Gessani S. HIV-1 gp120 activates the STAT3/interleukin-6 axis in primary human monocyte-derived dendritic cells. J Virol. 2014;88:11045–55.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Spear M, Guo J, Turner A, Yu D, Wang W, Meltzer B, et al. HIV-1 triggers WAVE2 phosphorylation in primary CD4 T cells and macrophages, mediating Arp2/3-dependent nuclear migration. J Biol Chem. 2014;289:6949–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Davis CB, Dikic I, Unutmaz D, Hill CM, Arthos J, Siani MA, et al. Signal transduction due to HIV-1 envelope interactions with chemokine receptors CXCR4 or CCR5. J Exp Med. 1997;186:1793–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Li L, Lim RZL, Lee LSU, Chew NSY. HIV glycoprotein gp120 enhances mesenchymal stem cell migration by upregulating CXCR4 expression. Biochim Et Biophys Acta Gen Subj. 2018;1862:1790–800.

    Article  CAS  Google Scholar 

  113. Shenoy SK, Lefkowitz RJ. β-arrestin-mediated receptor trafficking and signal transduction. Trends Pharmacol Sci. 2011;32:521–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kinter AL, Umscheid CA, Arthos J, Cicala C, Lin Y, Jackson R, et al. HIV envelope induces virus expression from resting CD4+ T cells isolated from HIV-infected individuals in the absence of markers of cellular activation or apoptosis. J Immunol. 2003;170:2449–55.

    Article  CAS  PubMed  Google Scholar 

  115. Juno JA, Fowke KR. Clarifying the role of G protein signaling in HIV infection: new approaches to an old question. AIDS Rev. 2010;12:164–76.

    PubMed  Google Scholar 

  116. Brandenberg OF, Magnus C, Regoes RR, Trkola A. The HIV-1 entry process: a stoichiometric view. Trends Microbiol. 2015;23:763–74.

    Article  CAS  PubMed  Google Scholar 

  117. Lin Y-L, Mettling C, Portales P, Reynes J, Clot J, Corbeau P. Cell surface CCR5 density determines the postentry efficiency of R5 HIV-1 infection. Proc Natl Acad Sci USA. 2002;99:15590–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lin Y-L, Mettling C, Portalès P, Réant B, Robert-Hebmann V, Reynes J, et al. The efficiency of R5 HIV-1 infection is determined by CD4 T-cell surface CCR5 density through Gαi-protein signalling. AIDS. 2006;20:1369–77.

    Article  CAS  PubMed  Google Scholar 

  119. Platt EJ, Wehrly K, Kuhmann SE, Chesebro B, Kabat D. Effects of CCR5 and CD4 cell surface concentrations on infections by Macrophagetropic isolates of human immunodeficiency virus type 1. J Virol. 1998;72:2855–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Reynes J, Portales P, Segondy M, Baillat V, André P, Réant B, et al. CD4+ T cell surface CCR5 density as a determining factor of virus load in persons infected with human immunodeficiency virus type 1. J Infect Dis. 2000;181:927–32.

    Article  CAS  PubMed  Google Scholar 

  121. Lelièvre J-D, Petit F, Perrin L, Mammano F, Arnoult D, Ameisen J-C, et al. The density of coreceptors at the surface of CD4+ T cells contributes to the extent of human immunodeficiency virus type 1 viral replication-mediated T cell death. AIDS Res Hum Retrovir. 2004;20:1230–43.

    PubMed  Google Scholar 

  122. Yang X, Jiao Y, Wang R, Ji Y, Zhang H, Zhang Y, et al. High CCR5 density on central memory CD4+ T cells in acute HIV-1 infection is mostly associated with rapid disease progression. PLoS ONE. 2012;7:e49526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Claireaux M, Robinot R, Kervevan J, Patgaonkar M, Staropoli I, Brelot A, et al. Low CCR5 expression protects HIV-specific CD4+ T cells of elite controllers from viral entry. Nat Commun. 2022;13:521.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Masquelier C, Servais J-Y, Rusanganwa E, Roman F, Havuga E, Servais J, et al. A novel 24-base pair deletion in the coding region of CCR5 in an African population. AIDS. 2007;21:111–3.

    Article  PubMed  Google Scholar 

  125. Arendt V, Amand M, Iserentant G, Lemaire M, Masquelier C, Ndayisaba GF, et al. Predominance of the heterozygous CCR5 delta-24 deletion in African individuals resistant to HIV infection might be related to a defect in CCR5 addressing at the cell surface. J Int AIDS Soc. 2019;22:e25384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. 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.

    Article  CAS  PubMed  Google Scholar 

  127. Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, Horuk R, et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell. 1996;86:367–77.

    Article  CAS  PubMed  Google Scholar 

  128. Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia growth and development study, multicenter AIDS cohort study, multicenter hemophilia cohort study, San Francisco city cohort, ALIVE study. Science. 1996;273:1856–62.

    Article  CAS  PubMed  Google Scholar 

  129. Stewart GJ, Ashton LJ, Biti RA, Ffrench RA, Bennetts BH, Newcombe NR, et al. Increased frequency of CCR-5 Δ32 heterozygotes among long-term non-progressors with HIV-1 infection. AIDS. 1997;11:1833–8.

    Article  CAS  PubMed  Google Scholar 

  130. Kuhmann SE, Platt EJ, Kozak SL, Kabat D. Cooperation of multiple CCR5 coreceptors is required for infections by human immunodeficiency virus type 1. J Virol. 2000;74:7005–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Baker A-M, Saulière A, Gaibelet G, Lagane B, Mazères S, Fourage M, et al. CD4 interacts constitutively with multiple CCR5 at the plasma membrane of living cells. A fluorescence recovery after photobleaching at variable radii approach. J Biol Chem. 2007;282:35163–8.

    Article  CAS  PubMed  Google Scholar 

  132. Wu L, Paxton WA, Kassam N, Ruffing N, Rottman JB, Sullivan N, et al. CCR5 levels and expression pattern correlate with infectability by macrophage-tropic HIV-1, in vitro. J Exp Med. 1997;185:1681–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bleul CC, Wu L, Hoxie JA, Springer TA, Mackay CR. The HIV coreceptors CXCR4 and CCR5 are differentially expressed and regulated on human T lymphocytes. Proc Natl Acad Sci USA. 1997;94:1925–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Joly M, Pinto JM. CXCR4 and CCR5 regulation and expression patterns on T- and monocyte-macrophage cell lineages: implications for susceptibility to infection by HIV-1. Math Biosci. 2005;195:92–126.

    Article  CAS  PubMed  Google Scholar 

  135. van Eekeren LE, Matzaraki V, Zhang Z, van de Wijer L, Blaauw MJT, de Jonge MI, et al. People with HIV have higher percentages of circulating CCR5+ CD8+ T cells and lower percentages of CCR5+ regulatory T cells. Sci Rep. 2022;12:11425.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Dale NC, Johnstone EKM, Pfleger KDG. GPCR heteromers: an overview of their classification, function and physiological relevance. Front Endocrinol. 2022;13:931573.

    Article  Google Scholar 

  137. Pin J-P, Neubig R, Bouvier M, Devi L, Filizola M, Javitch JA, et al. International union of basic and clinical pharmacology. LXVII. Recommendations for the recognition and nomenclature of G protein-coupled receptor heteromultimers. Pharmacol Rev. 2007;59:5–13.

    Article  CAS  PubMed  Google Scholar 

  138. Ferré S, Baler R, Bouvier M, Caron MG, Devi LA, Durroux T, et al. Building a new conceptual framework for receptor heteromers. Nat Chem Biol. 2009;5:131–4.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Muñoz LM, Lucas P, Holgado BL, Barroso R, Vega B, Rodríguez-Frade JM, et al. Receptor oligomerization: a pivotal mechanism for regulating chemokine function. Pharmacol Ther. 2011;131:351–8.

    Article  PubMed  Google Scholar 

  140. Stephens B, Handel TM. Chemokine receptor oligomerization and allostery. Prog Mol Biol Transl Sci. 2013;115:375–420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Martínez-Muñoz L, Villares R, Rodríguez-Fernández JL, Rodríguez-Frade JM, Mellado M. Remodeling our concept of chemokine receptor function: from monomers to oligomers. J Leukoc Biol. 2018;104:323–31.

    Article  PubMed  Google Scholar 

  142. Issafras H, Angers S, Bulenger S, Blanpain C, Parmentier M, Labbé-Jullié C, et al. Constitutive agonist-independent CCR5 oligomerization and antibody-mediated clustering occurring at physiological levels of receptors. J Biol Chem. 2002;277:34666–73.

    Article  CAS  PubMed  Google Scholar 

  143. Nakano Y, Monde K, Terasawa H, Yuan Y, Yusa K, Harada S, et al. Preferential recognition of monomeric CCR5 expressed in cultured cells by the HIV-1 envelope glycoprotein gp120 for the entry of R5 HIV-1. Virology. 2014;452–453:117–24.

    Article  PubMed  Google Scholar 

  144. Li J, Ding Y, Liu H, He H, Yu D, Wang X, et al. Oligomerization-enhanced receptor-ligand binding revealed by dual-color simultaneous tracking on living cell membranes. J Phys Chem Lett. 2021;12:8164–9.

    Article  CAS  PubMed  Google Scholar 

  145. Momboisse F, Nardi G, Colin P, Hery M, Cordeiro N, Blachier S, et al. Tracking receptor motions at the plasma membrane reveals distinct effects of ligands on CCR5 dynamics depending on its dimerization status. Elife. 2022;11:e76281.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Jin J, Momboisse F, Boncompain G, Koensgen F, Zhou Z, Cordeiro N, et al. CCR5 adopts three homodimeric conformations that control cell surface delivery. Sci Signal. 2018;11:l2869.

    Article  Google Scholar 

  147. El-Asmar L, Springael J-Y, Ballet S, Andrieu EU, Vassart G, Parmentier M. Evidence for negative binding cooperativity within CCR5-CCR2b heterodimers. Mol Pharmacol. 2005;67:460–9.

    Article  CAS  PubMed  Google Scholar 

  148. Springael J-Y, 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.

    Article  CAS  PubMed  Google Scholar 

  149. Martínez-Muñoz L, Barroso R, Dyrhaug SY, Navarro G, Lucas P, Soriano SF, et al. CCR5/CD4/CXCR4 oligomerization prevents HIV-1 gp120IIIB binding to the cell surface. Proc Natl Acad Sci USA. 2014;111:E1960-1969.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Möller J, Isbilir A, Sungkaworn T, Osberg B, Karathanasis C, Sunkara V, et al. Single-molecule analysis reveals agonist-specific dimer formation of µ-opioid receptors. Nat Chem Biol. 2020;16:946–54.

    Article  PubMed  Google Scholar 

  151. Tabor A, Weisenburger S, Banerjee A, Purkayastha N, Kaindl JM, Hübner H, et al. Visualization and ligand-induced modulation of dopamine receptor dimerization at the single molecule level. Sci Rep. 2016;6:33233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Laganowsky A, Reading E, Allison TM, Ulmschneider MB, Degiacomi MT, Baldwin AJ, et al. Membrane proteins bind lipids selectively to modulate their structure and function. Nature. 2014;510:172–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Levental I, Lyman E. Regulation of membrane protein structure and function by their lipid nano-environment. Nat Rev Mol Cell Biol. 2022. https://doi.org/10.1038/s41580-022-00524-4.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Dawaliby R, Trubbia C, Delporte C, Masureel M, Van Antwerpen P, Kobilka BK, et al. Allosteric regulation of G protein-coupled receptor activity by phospholipids. Nat Chem Biol. 2016;12:35–9.

    Article  CAS  PubMed  Google Scholar 

  155. Jakubík J, El-Fakahany EE. Allosteric modulation of GPCRs of class A by cholesterol. Int J Mol Sci. 2021;22:1953.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Oates J, Watts A. Uncovering the intimate relationship between lipids, cholesterol and GPCR activation. Curr Opin Struct Biol. 2011;21:802–7.

    Article  CAS  PubMed  Google Scholar 

  157. Calmet P, Cullin C, Cortès S, Vang M, Caudy N, Baccouch R, et al. Cholesterol impacts chemokine CCR5 receptor ligand-binding activity. FEBS J. 2020;287:2367–85.

    Article  CAS  PubMed  Google Scholar 

  158. Cardaba CM, Kerr JS, Mueller A. CCR5 internalisation and signalling have different dependence on membrane lipid raft integrity. Cell Signal. 2008;20:1687–94.

    Article  CAS  PubMed  Google Scholar 

  159. Ablan S, Rawat SS, Viard M, Wang JM, Puri A, Blumenthal R. The role of cholesterol and sphingolipids in chemokine receptor function and HIV-1 envelope glycoprotein-mediated fusion. Virol J. 2006;3:104.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Gardeta SR, García-Cuesta EM, D’Agostino G, Soler Palacios B, Quijada-Freire A, Lucas P, et al. Sphingomyelin depletion inhibits CXCR4 dynamics and CXCL12-mediated directed cell migration in human T cells. Front Immunol. 2022. https://doi.org/10.3389/fimmu.2022.925559.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Denard B, Han S, Kim J, Ross EM, Ye J. Regulating G protein-coupled receptors by topological inversion. Elife. 2019;8:e40234.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Castro BM, Prieto M, Silva LC. Ceramide: a simple sphingolipid with unique biophysical properties. Prog Lipid Res. 2014;54:53–67.

    Article  CAS  PubMed  Google Scholar 

  163. Simons K, Sampaio JL. Membrane organization and lipid rafts. Cold Spring Harb Perspect Biol. 2011;3:a004697.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Chazal N, Gerlier D. Virus entry, assembly, budding, and membrane rafts. Microbiol Mol Biol Rev. 2003;67:226–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Parolini I, Topa S, Sorice M, Pace A, Ceddia P, Montesoro E, et al. Phorbol ester-induced disruption of the CD4-Lck complex occurs within a detergent-resistant microdomain of the plasma membrane. Involvement of the translocation of activated protein kinase C isoforms. J Biol Chem. 1999;274:14176–87.

    Article  CAS  PubMed  Google Scholar 

  166. Millán J, Cerny J, Horejsi V, Alonso MA. CD4 segregates into specific detergent-resistant T-cell membrane microdomains. Tissue Antigens. 1999;53:33–40.

    Article  PubMed  Google Scholar 

  167. Gaibelet G, Planchenault T, Mazères S, Dumas F, Arenzana-Seisdedos F, Lopez A, et al. CD4 and CCR5 constitutively interact at the plasma membrane of living cells: a confocal fluorescence resonance energy transfer-based approach. J Biol Chem. 2006;281:37921–9.

    Article  CAS  PubMed  Google Scholar 

  168. Popik W, Alce TM, Au W-C. Human immunodeficiency virus type 1 uses lipid raft-colocalized CD4 and chemokine receptors for productive entry into CD4(+) T cells. J Virol. 2002;76:4709–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Kozak SL, Heard JM, Kabat D. Segregation of CD4 and CXCR4 into distinct lipid microdomains in T lymphocytes suggests a mechanism for membrane destabilization by human immunodeficiency virus. J Virol. 2002;76:1802–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Kamiyama H, Yoshii H, Tanaka Y, Sato H, Yamamoto N, Kubo Y. Raft localization of CXCR4 is primarily required for X4-tropic human immunodeficiency virus type 1 infection. Virology. 2009;386:23–31.

    Article  CAS  PubMed  Google Scholar 

  171. Nguyen DH, Taub D. Cholesterol is essential for macrophage inflammatory protein 1 beta binding and conformational integrity of CC chemokine receptor 5. Blood. 2002;99:4298–306.

    Article  CAS  PubMed  Google Scholar 

  172. Yang S-T, Kiessling V, Simmons JA, White JM, Tamm LK. HIV gp41-mediated membrane fusion occurs at edges of cholesterol-rich lipid domains. Nat Chem Biol. 2015;11:424–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Yang S-T, Kiessling V, Tamm LK. Line tension at lipid phase boundaries as driving force for HIV fusion peptide-mediated fusion. Nat Commun. 2016;7:11401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Solloch UV, Lang K, Lange V, Böhme I, Schmidt AH, Sauter J. Frequencies of gene variant CCR5-Δ32 in 87 countries based on next-generation sequencing of 1.3 million individuals sampled from 3 national DKMS donor centers. Human Immunol. 2017;78:710–7.

    Article  CAS  Google Scholar 

  175. Benkirane M, Jin DY, Chun RF, Koup RA, Jeang KT. Mechanism of transdominant inhibition of CCR5-mediated HIV-1 infection by ccr5delta32. J Biol Chem. 1997;272:30603–6.

    Article  CAS  PubMed  Google Scholar 

  176. Agrawal L, Lu X, Qingwen J, VanHorn-Ali Z, Nicolescu IV, McDermott DH, et al. Role for CCR5Delta32 protein in resistance to R5, R5X4, and X4 human immunodeficiency virus type 1 in primary CD4+ cells. J Virol. 2004;78:2277–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 2009;360:692–8.

    Article  PubMed  Google Scholar 

  178. Gupta RK, Abdul-jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem cell transplantation. Nature. 2019;568:244–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Ding J, Liu Y, Lai Y. Knowledge from London and Berlin: finding threads to a functional HIV cure. Front Immunol. 2021;12:688747.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Hsu J, Van Besien K, Glesby MJ, Coletti A, Pahwa SG, Warshaw M, et al. Hiv-1 Remission With Ccr5∆32∆32 Haplo-Cord Transplant In A U .S . Woman:IMPAACT P1107. Abstracts From CROI 2022 Conference on Retroviruses and Opportunistic Infections CROI 2022 Abstract eBook 2022. :26.

  181. Jensen B-EO, Knops E, Cords L, Lübke N, Salgado M, Busman-Sahay K, et al. In-depth virological and immunological characterization of HIV-1 cure after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation. Nat Med. 2023. https://doi.org/10.1038/s41591-023-02213-x.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Fischereder M, Luckow B, Hocher B, Wüthrich RP, Rothenpieler U, Schneeberger H, et al. CC chemokine receptor 5 and renal-transplant survival. The Lancet. 2001;357:1758–61.

    Article  CAS  Google Scholar 

  183. Ferrarini I, Rigo A, Gandini A, Vinante F. West nile virus encephalitis in haematological setting: report of two cases and a brief review of the literature. Mediterr J Hematol Infect Dis. 2019;11:e2019033.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Baba M, Takashima K, Miyake H, Kanzaki N, Teshima K, Wang X, et al. TAK-652 inhibits CCR5-mediated human immunodeficiency virus type 1 infection in vitro and has favorable pharmacokinetics in humans. Antimicrob Agents Chemother. 2005;49:4584–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Wolinsky SM, Veazey RS, Kunstman KJ, Klasse PJ, Dufour J, Marozsan AJ, et al. Effect of a CCR5 inhibitor on viral loads in macaques dual-infected with R5 and X4 primate immunodeficiency viruses. Virology. 2004;328:19–29.

    Article  CAS  PubMed  Google Scholar 

  186. Maeda K, Nakata H, Koh Y, Miyakawa T, Ogata H, Takaoka Y, et al. Spirodiketopiperazine-based CCR5 inhibitor which preserves CC-chemokine/CCR5 interactions and exerts potent activity against R5 human immunodeficiency virus type 1 in vitro. J Virol. 2004;78:8654–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Kim MB, Giesler KE, Tahirovic YA, Truax VM, Liotta DC, Wilson LJ. CCR5 receptor antagonists in preclinical to phase II clinical development for treatment of HIV. Expert Opin Investig Drugs. 2016;25:1377–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Biswas P, Tambussi G, Lazzarin A. Access denied? The status of co-receptor inhibition to counter HIV entry. Expert Opin Pharmacother. 2007;8:923–33.

    Article  CAS  PubMed  Google Scholar 

  189. Kuritzkes D, Kar S, Kirkpatrick P. Maraviroc. Nat Rev Drug Discovery. 2008;7:15–6.

    Article  CAS  Google Scholar 

  190. Lagane B, Garcia-Perez J, Kellenberger E. Modeling the allosteric modulation of CCR5 function by Maraviroc. Drug Discov Today Technol. 2013;10:e297-305.

    Article  PubMed  Google Scholar 

  191. Garcia-Perez J, Rueda P, Alcami J, Rognan D, Arenzana-Seisdedos F, Lagane B, et al. Allosteric model of maraviroc binding to CC chemokine receptor 5 (CCR5). J Biol Chem. 2011;286:33409–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Moore JP, Kuritzkes DR. A pièce de resistance: how HIV-1 escapes small molecule CCR5 inhibitors. Curr Opin HIV AIDS. 2009;4:118–24.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Westby M, Smith-Burchnell C, Mori J, Lewis M, Mosley M, Stockdale M, et al. Reduced maximal inhibition in phenotypic susceptibility assays indicates that viral strains resistant to the CCR5 antagonist maraviroc utilize inhibitor-bound receptor for entry. J Virol. 2007;81:2359–71.

    Article  CAS  PubMed  Google Scholar 

  194. Pugach P, Marozsan AJ, Ketas TJ, Landes EL, Moore JP, Kuhmann SE. HIV-1 clones resistant to a small molecule CCR5 inhibitor use the inhibitor-bound form of CCR5 for entry. Virology. 2007;361:212–28.

    Article  CAS  PubMed  Google Scholar 

  195. Promsote W, DeMouth ME, Almasri CG, Pegu A. Anti-HIV-1 antibodies: an update. BioDrugs. 2020;34:121–32.

    Article  CAS  PubMed  Google Scholar 

  196. Li L, Sun T, Yang K, Zhang P, Jia WQ. Monoclonal CCR5 antibody for treatment of people with HIV infection. Cochrane Database Syst Rev. 2010. https://doi.org/10.1002/14651858.CD008439.pub2.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Lalezari J, Yadavalli GK, Para M, Richmond G, Dejesus E, Brown SJ, et al. Safety, pharmacokinetics, and antiviral activity of HGS004, a novel fully human IgG4 monoclonal antibody against CCR5, in HIV-1-infected patients. J Infect Dis. 2008;197:721–7.

    Article  CAS  PubMed  Google Scholar 

  198. Venuti A, Pastori C, Lopalco L. The role of natural antibodies to CC chemokine receptor 5 in HIV infection. Front Immunol. 2017;8:1358.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Kufel WD. Antibody-based strategies in HIV therapy. Int J Antimicrob Agents. 2020;56:106186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Jacobson JM, Thompson MA, Lalezari JP, Saag MS, Zingman P, D’Ambrosio BS, et al. Anti-HIV-1 activity of weekly or biweekly treatment with subcutaneous PRO 140, a CCR5 monoclonal antibody. J Infect Dis. 2010;201:1481–7.

    Article  CAS  PubMed  Google Scholar 

  201. Jacobson JM, Lalezari JP, Thompson MA, Fichtenbaum CJ, Saag MS, Zingman BS, et al. Phase 2a study of the CCR5 monoclonal antibody PRO 140 administered intravenously to HIV-infected adults. Antimicrob Agents Chemother. 2010;54:4137–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Pfeifer N, Walter H, Lengauer T. Association between HIV-1 coreceptor usage and resistance to broadly neutralizing antibodies. J Acquir Immune Defic Syndr. 2014;67:107–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Lu Y, Happi Mbakam C, Song B, Bendavid E, Tremblay J-P. Improvements of nuclease and nickase gene modification techniques for the treatment of genetic diseases. Front Genome Ed. 2022;4:892769.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Freen-van Heeren JJ. Closing the door with CRISPR: genome editing of CCR5 and CXCR4 as a potential curative solution for HIV. BioTech. 2022;11:25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Bhowmik R, Chaubey B. CRISPR/Cas9: a tool to eradicate HIV-1. AIDS Res Ther. 2022;19:58.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Wang W, Ye C, Liu J, Zhang D, Kimata JT, Zhou P. CCR5 gene disruption via lentiviral vectors expressing Cas9 and single guided RNA renders cells resistant to HIV-1 infection. PLoS ONE. 2014;9:e115987.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Tebas P, Jadlowsky JK, Shaw PA, Tian L, Esparza E, Brennan AL, et al. CCR5-edited CD4+ T cells augment HIV-specific immunity to enable post-rebound control of HIV replication. J Clin Invest. 2021;131(e144486):144486.

    Article  PubMed  Google Scholar 

  209. Herskovitz J, Hasan M, Patel M, Blomberg WR, Cohen JD, Machhi J, et al. CRISPR-Cas9 mediated exonic disruption for HIV-1 elimination. EBioMedicine. 2021;73:103678.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, et al. CRISPR-edited stem cells in a patient with HIV and acute lymphocytic leukemia. N Engl J Med. 2019;381:1240–7.

    Article  CAS  PubMed  Google Scholar 

  211. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370:901–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Cho SW, Kim S, Kim JM, Kim J-S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol. 2013;31:230–2.

    Article  CAS  PubMed  Google Scholar 

  213. Cradick TJ, Fine EJ, Antico CJ, Bao G. CRISPR/Cas9 systems targeting β-globin and CCR5 genes have substantial off-target activity. Nucleic Acids Res. 2013;41:9584–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Li S, Holguin L, Burnett JC. CRISPR-Cas9-mediated gene disruption of HIV-1 co-receptors confers broad resistance to infection in human T cells and humanized mice. Mol Ther Methods Clin Dev. 2022;24:321–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, et al. Seamless modification of wild-type induced pluripotent stem cells to the natural CCR5Δ32 mutation confers resistance to HIV infection. Proc Natl Acad Sci. 2014;111:9591–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Qi C, Li D, Jiang X, Jia X, Lu L, Wang Y, et al. Inducing CCR5Δ32/Δ32 homozygotes in the human jurkat CD4+ cell line and primary CD4+ Cells by CRISPR-Cas9 genome-editing technology. Mol Ther Nucl Acids. 2018;12:267–74.

    Article  Google Scholar 

  217. Jasinska AJ, Pandrea I, Apetrei C. CCR5 as a coreceptor for human immunodeficiency virus and simian immunodeficiency viruses: a prototypic love-hate affair. Front Immunol. 2022;13:835994.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Jacquemard C, Koensgen F, Colin P, Lagane B, Kellenberger E. Modeling of CCR5 recognition by HIV-1 gp120: how the viral protein exploits the conformational plasticity of the coreceptor. Viruses. 2021;13:1395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Katritch V, Cherezov V, Stevens RC. Diversity and modularity of G protein-coupled receptor structures. Trends Pharmacol Sci. 2012;33:17–27.

    Article  CAS  PubMed  Google Scholar 

  220. Weichseldorfer M, Tagaya Y, Reitz M, DeVico AL, Latinovic OS. Identifying CCR5 coreceptor populations permissive for HIV-1 entry and productive infection: implications for in vivo studies. J Transl Med. 2022;20:39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Abrol R, Trzaskowski B, Goddard WA, Nesterov A, Olave I, Irons C. Ligand- and mutation-induced conformational selection in the CCR5 chemokine G protein-coupled receptor. Proc Natl Acad Sci USA. 2014;111:13040–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Lee B, Sharron M, Blanpain C, Doranz BJ, Vakili J, Setoh P, et al. Epitope mapping of CCR5 reveals multiple conformational states and distinct but overlapping structures involved in chemokine and coreceptor function. J Biol Chem. 1999;274:9617–26.

    Article  CAS  PubMed  Google Scholar 

  223. Kleist AB, Getschman AE, Ziarek JJ, Nevins AM, Gauthier P-A, Chevigné A, et al. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem Pharmacol. 2016;114:53–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Juremalm M, Nilsson G. Chemokine receptor expression by mast cells. Chem Immunol Allergy. 2005;87:130–44.

    Article  CAS  PubMed  Google Scholar 

  225. Rottman JB, Ganley KP, Williams K, Wu L, Mackay CR, Ringler DJ. Cellular localization of the chemokine receptor CCR5. Correlation to cellular targets of HIV-1 infection. Am J Pathol. 1997;151:1341–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Fox JM, Kasprowicz R, Hartley O, Signoret N. CCR5 susceptibility to ligand-mediated down-modulation differs between human T lymphocytes and myeloid cells. J Leukoc Biol. 2015;98:59–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Lagane B, Ballet S, Planchenault T, Balabanian K, Le Poul E, Blanpain C, et al. Mutation of the DRY motif reveals different structural requirements for the CC chemokine receptor 5-mediated signaling and receptor endocytosis. Mol Pharmacol. 2005;67:1966–76.

    Article  CAS  PubMed  Google Scholar 

  228. Jin J, Colin P, Staropoli I, Lima-Fernandes E, Ferret C, Demir A, et al. Targeting spare CC chemokine receptor 5 (CCR5) as a principle to inhibit HIV-1 entry. J Biol Chem. 2014;289:19042–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Armani-Tourret M, Zhou Z, Gasser R, Staropoli I, Cantaloube-Ferrieu V, Benureau Y, et al. Mechanisms of HIV-1 evasion to the antiviral activity of chemokine CXCL12 indicate potential links with pathogenesis. PLoS Pathog. 2021;17:e1009526.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Zheng Y, Han GW, Abagyan R, Wu B, Stevens RC, Cherezov V, et al. Structure of CC chemokine receptor 5 with a potent chemokine antagonist reveals mechanisms of chemokine recognition and molecular mimicry by HIV. Immunity. 2017;46:1005-1017.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Hemelaar J. Implications of HIV diversity for the HIV-1 pandemic. J Infect. 2013;66:391–400.

    Article  PubMed  Google Scholar 

  232. Tee KK, Thomson MM, Hemelaar J. Editorial: HIV-1 genetic diversity, volume II. Front Microbiol. 2022;13:1007037.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Kawashima Y, Pfafferott K, Frater J, Matthews P, Payne R, Addo M, et al. Adaptation of HIV-1 to human leukocyte antigen class I. Nature. 2009;458:641–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Chawla A, Wang C, Patton C, Murray M, Punekar Y, de Ruiter A, et al. A review of long-term toxicity of antiretroviral treatment regimens and implications for an aging population. Infect Dis Ther. 2018;7:183–95.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Lopalco L. Natural anti-CCR5 antibodies in HIV-infection and -exposure. J Transl Med. 2011;9:S4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Yoo KD, Cha R, Lee S, Kim JE, Kim KH, Lee JS, et al. Chemokine receptor 5 blockade modulates macrophage trafficking in renal ischaemic-reperfusion injury. J Cell Mol Med. 2020;24:5515–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the Centre National de la Recherche Scientifique, Université Paul Sabatier, the Institut National de la Santé et de la Recherche Médicale, Sidaction (13457) and the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS) grant numbers: ANRS2018-02, ECTZ 118551/118554, ECTZ 205320/305352, ANRS ECTZ103104 and ECTZ101971. N.F is a PhD candidate at Université Toulouse III Paul Sabatier supported by ANRS.

Author information

Authors and Affiliations

Authors

Contributions

All authors equally contributed to the redaction of this mini-review. The authors thanks the reviewers for their constructive suggestions that contributed to improve the manuscript.

Corresponding author

Correspondence to Fabrice Dumas.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no scientific, commercial or financial competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Faivre, N., Verollet, C. & Dumas, F. The chemokine receptor CCR5: multi-faceted hook for HIV-1. Retrovirology 21, 2 (2024). https://doi.org/10.1186/s12977-024-00634-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12977-024-00634-1

Keywords