Next Article in Journal
Gender Medicine and Pharmacology
Next Article in Special Issue
Neuropathic Pain and Related Depression in Mice: The Effect of a Terpene and a Minor Cannabinoid in Combination
Previous Article in Journal
Selective Cytotoxicity in Chronic Myeloid Leukemia (K-562) Cells Induced by 532 nm LASER Irradiation Without Exogenous Photosensitizers
Previous Article in Special Issue
Genetic, Psychological, and Behavioural Factors Associated with Subtypes of Pain-Related Temporomandibular Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the CCR5 Receptor in Neuropathic Pain Modulation: Current Insights and Therapeutic Implications

by
Mario García-Domínguez
1,2,3
1
Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain
2
Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
3
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
Biomedicines 2025, 13(11), 2650; https://doi.org/10.3390/biomedicines13112650
Submission received: 24 September 2025 / Revised: 20 October 2025 / Accepted: 22 October 2025 / Published: 29 October 2025
(This article belongs to the Special Issue Biomarkers in Pain: 2nd Edition)

Abstract

Neuropathic pain, a chronic condition arising from injury or dysfunction of the somatosensory nervous system, is characterized by persistent hypersensitivity and spontaneous pain. The chemokine receptor CCR5 (C-C motif chemokine receptor 5) has recently been identified as a critical mediator in neuroinflammation and neuropathic pain signaling pathways. Expressed on immune cells and neurons, CCR5 regulates immune cell recruitment and activation, thereby contributing to neuronal sensitization and maintenance of pain states. This review examines the currently characterized molecular mechanisms through which CCR5 modulates neuropathic pain pathophysiology and assesses the potential of CCR5 antagonists as novel therapeutic agents for the management of chronic neuropathic pain. Understanding the involvement of CCR5 in pain modulation may facilitate the development of targeted treatments with improved efficacy and safety profiles.

1. Introduction

The International Association for the Study of Pain (IASP) defines pain as an “unpleasant sensory and emotional experience associated with, or resembling that associated with, actual or potential tissue damage” [1,2]. This definition highlights that pain is a multifactorial phenomenon that serves as a critical diagnostic indicator, frequently signaling underlying health conditions that require medical intervention [3]. Pain can be classified into distinct categories according to its underlying characteristics, including its origin [4]. Nociceptive pain arises from actual or potential damage to non-neural tissue due to the activation of nociceptors [5]. Nociplastic pain originates from altered nociception in the absence of clear evidence of actual or potential tissue injury that would activate peripheral nociceptors, or without any demonstrable disease or lesion of the somatosensory system capable of causing pain [6]. Finally, neuropathic pain results from a lesion or disease affecting the somatosensory nervous system; however, this pain subtype will be addressed in detail later.
Neuropathic pain is a highly prevalent, chronic, and debilitating condition that arises as a consequence of injury or dysfunction within the somatosensory nervous system [7]. It represents a significant global health burden, affecting an estimated 7–10% of the population, and is associated with reduced quality of life, impaired daily functioning, and substantial socioeconomic costs [8,9,10]. Unlike nociceptive pain, which occurs from actual or potential tissue damage and serves as a protective mechanism by signaling the presence of noxious stimuli, neuropathic pain occurs independently of peripheral injury and frequently manifests as a maladaptive, chronic condition [11,12].
Clinically, neuropathic pain presents with a heterogeneous spectrum of sensory abnormalities. Patients usually report spontaneous pain that occurs without any apparent external trigger, which might manifest as continuous burning, tingling, or aching sensations [13]. In addition, paroxysmal episodes of sharp, shooting, or electric shock-like pain are often observed, reflecting aberrant excitability within nociceptive pathways [14]. Neuropathic pain is also associated with increased responsiveness to noxious stimuli, referred to as hyperalgesia, as well as the induction of pain by normally innocuous stimuli, known as allodynia [15,16]. These sensory disturbances can be further classified into mechanical, thermal, or dynamic subtypes, underscoring the complex and multifaceted nature of the disorder and its impact on somatosensory processing [17]. The complex symptomatology and interindividual variability in clinical presentation highlight the multifactorial nature of neuropathic pain and the need for a deeper understanding of its molecular underpinnings [18].
From a molecular perspective, neuropathic pain is increasingly understood as more than aberrant neuronal firing; it represents a complex neuroimmune disorder in which bidirectional communication between immune cells, glial cells, and neurons orchestrates changes in neuronal excitability and synaptic architecture [19]. Peripheral nerve injury or CNS insult rapidly triggers a cascade of inflammatory and immune responses. Damaged neurons, Schwann cells, and surrounding non-neuronal cells release many pro-inflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6), chemokines (e.g., CCL2, CCL3, and CCL5), and damage-associated molecular patterns (DAMPs) [20,21,22]. These molecules operate as danger signals, activating resident glial populations (primarily microglia and astrocytes) via pattern recognition (PRRs) and cytokine/chemokine receptors [23,24,25]. Activated glial cells, in turn, release additional pro-inflammatory mediators and neurotrophic factors that modulate neuronal activity and synaptic function (such as IL-1β, TNF-α, BDNF, and NGF) [26,27]. In parallel, chemokine gradients and adhesion molecules (e.g., ICAM-1) foster the recruitment and infiltration of peripheral immune cells, including macrophages, T lymphocytes, and neutrophils, into the injured nerve and central pain-processing regions including the spinal dorsal horn and supraspinal centers [21,28]. This long-lasting neuroinflammatory environment promotes profound alterations in neuronal physiology. VGSCs (Nav1.3 and Nav1.7), VGCCs (e.g., Cav2.2), and K+ channels show altered expression and biophysical properties, contributing to ectopic firing and hyperexcitability of nociceptive neurons [29,30,31]. Simultaneously, dysregulation of excitatory neurotransmitters such as glutamate and aspartate, along with impaired inhibitory GABAergic and glycinergic signaling, facilitates central sensitization [32,33]. Maladaptive synaptic plasticity, including long-term potentiation (LTP) at dorsal horn synapses, amplifies nociceptive signal transmission and underlies persistent hypersensitivity to both noxious and innocuous stimuli [34].
Among the diverse molecular mediators that orchestrate neuroimmune communication in neuropathic pain, chemokine signaling has emerged as a key determinant of pain hypersensitivity [35]. Chemokines, a highly conserved family of small secreted cytokines (8–12 kDa), are classified into four major subfamilies (CC, CXC, CX3C, and XC) based on the arrangement of conserved cysteine residues, with receptors such as CCR1-CCR10, CXCR1-CXCR7, CX3CR1, and XCR1 mediating their biological effects [36]. These proteins exert numerous roles in the immune and nervous systems, as they modulate the recruitment, migration, and activation of immune cells (e.g., microglia, macrophages, and T cells) at sites of neural injury or inflammation [37]. Beyond their classical immunomodulatory functions, chemokines influence neuronal activity by modulating membrane excitability, synaptic transmission, and the plasticity of pain pathways [38].
The CCR5 receptor has garnered considerable attention for its involvement in regulating immune responses and modulating neuronal excitability [39,40]. CCR5 is a GPCR expressed across diverse subsets of immune cells (such as macrophages, monocytes, and T lymphocytes) [41], along with glial cells (such as microglia and astrocytes) [42,43], and some neuronal populations [44]. Upon binding its endogenous ligands, such as CCL3, CCL4, and CCL5, CCR5 activates several signaling cascades, in particular, the PI3K/Akt, MAPK/ERK, and NF-κB pathways [45,46]. Activation of these pathways controls the recruitment of additional immune cells, boosts glial reactivity, promotes transcription and release of secondary pro-nociceptive mediators (e.g., IL-1β), and supports synaptic modifications in dorsal horn neurons [47]. Collectively, these processes establish a self-perpetuating feed-forward loop of neuroinflammation, glial activation, and neuronal sensitization that drives the initiation and chronicity of neuropathic pain [48,49].
Despite decades of research, current therapeutic options for neuropathic pain remain unsatisfactory. Conventional first-line therapies, such as anticonvulsants (e.g., gabapentinoids) and antidepressants (e.g., amitriptyline), produce limited analgesic efficacy in a proportion of patients [50,51]. Opioids, although usually prescribed, are characterized by limited effectiveness, the propensity for tolerance induction, and an increased risk of dependence [52]. Moreover, several available drugs carry substantial adverse effects, involving sedation, dizziness, gastrointestinal disturbances, and cognitive impairment, which further restrict their long-term utility [53]. These limitations underscore the need for novel therapeutic strategies with improved efficacy that are mechanism-oriented, targeting the cellular and molecular pathways responsible for the maintenance of neuropathic pain.
In the context of neuropathic pain, CCR5 is recognized to exert dual effects on pain regulation (mediating immune responses and influencing neuronal excitability). This role of CCR5 establishes it as a promising therapeutic target, bridging immune and neuronal pathways that underlie chronic pain [54]. Pharmacological blockade or genetic deletion of CCR5 has been shown in experimental models to attenuate glial activation, reduce cytokine and chemokine production, normalize neuronal excitability, and ultimately alleviate pain-like behaviors [55,56]. Indeed, emerging evidence increasingly implicates CCR5 as a pivotal regulator of neuroinflammatory pathways involved in the initiation and maintenance of neuropathic pain. Preclinical rodent models of neuropathic pain employ diverse strategies to mimic distinct clinical phenotypes. Chronic constriction injury (CCI) involves loose ligation of the sciatic nerve, inducing partial nerve damage and persistent mechanical and thermal hypersensitivity [57]. Partial sciatic nerve ligation (PSNL) consists of tight ligation of a segment of the sciatic nerve, producing robust and long-lasting allodynia and hyperalgesia [57]. Chemotherapy-induced peripheral neuropathy (CIPN) is generated by administration of neurotoxic chemotherapeutics, such as paclitaxel or oxaliplatin, causing sensory neuropathy with mechanical and thermal hypersensitivity [58]. Diabetic neuropathy (DPN) is modeled via streptozotocin injection or genetic diabetes, leading to chronic hyperglycemia and progressive sensory deficits, recapitulating key features of human diabetic neuropathy [59]. In view of the convergence of immune and neuronal signaling at the level of CCR5, targeted inhibition of this receptor holds considerable promise for the development of novel mechanism-based therapies for neuropathic pain.
This review provides a comprehensive overview of the molecular and cellular mechanisms through which CCR5 contributes to the initiation and maintenance of neuropathic pain. In addition, the therapeutic potential of CCR5 antagonists (many of which have been developed and evaluated in other clinical contexts, such as HIV-1 infection) is assessed as a strategy for the treatment of chronic pain.

2. CCR5: Structure and Physiological Functions

2.1. Structural Features and Molecular Signaling Mechanisms of CCR5

The CCR5 is a prototypical member of the class A GPCR family, a large and evolutionarily conserved group of membrane proteins that mediate cell communication in response to extracellular stimuli [60]. Similarly to other G protein-coupled receptors, CCR5 is characterized by seven transmembrane (7TM) α-helical domains organized in a canonical serpentine topology that crosses the lipid bilayer [61]. These helices are interconnected by three extracellular loops (ECL1–ECL3) and three intracellular loops (ICL1–ICL3), creating a structural framework that allows dynamic conformational rearrangements upon ligand binding [62].
At the extracellular level, CCR5 possesses an N-terminal domain rich in tyrosine residues that are frequently sulfated [63], a post-translational modification essential for high-affinity binding to several chemokine ligands, including CCL3, CCL4, and CCL5. In addition, this N-terminus undergoes O- and N-linked glycosylation, which modulates receptor folding, trafficking to the plasma membrane, and ligand specificity [64,65]. The extracellular loops, particularly ECL2, also contribute critically to chemokine recognition and fine-tuning of ligand selectivity, forming a multi-point binding interface that coordinates the chemokine’s core domain with the receptor pocket [66].
The transmembrane helices assemble into a ligand-binding pocket that represents the structural core of the receptor’s activation mechanism. Upon chemokine engagement, local rearrangements of the 7TM bundle are transmitted inward, especially affecting TM3, TM5, and TM6, which act as “microswitches” that propagate the signal toward intracellular domains [67]. These conformational changes enable the intracellular regions of CCR5 to interact with heterotrimeric G proteins of the Gi/q families, leading to the dissociation of Gαi/q from the Gβγ subunits [68]. This dissociation triggers some signaling events (Figure 1): inhibition of adenylyl cyclase activity (reducing intracellular cAMP levels) [69], activation of phospholipase C-β (PLC-β) with subsequent hydrolysis of PIP2 into IP3 and DAG, mobilization of Ca2+ from intracellular stores [70], and activation of protein kinase C (PKC) [71]. In parallel, other signaling routes upregulate the MAPK/ERK cascade, the PI3K/Akt pathway, and members of the Rho family of small GTPases, which collectively orchestrate some cellular processes such as migration, adhesion, survival, and transcriptional regulation [72,73,74].
The intracellular C-terminal tail of CCR5 is enriched in serine and threonine residues that serve as phosphorylation sites for GRKs [75]. Following phosphorylation, these residues recruit β-arrestins, multifunctional adaptor proteins that not only promote receptor internalization through clathrin-coated pits but also with scaffold alternative, G protein-independent signaling pathways [76]. Through this dual role, β-arrestins contribute both to signal desensitization (terminating G protein activation) and to the initiation of noncanonical signaling cascades, including those leading to ERK1/2 activation [77]. CCR5 transport among the plasma membrane, endosomes, and recycling compartments is stringently regulated to balance responsiveness to extracellular chemokines with receptor availability at the cell surface [78].
In terms of evolution and genetics, CCR5 is conserved among vertebrates, underscoring its key role in immune system regulation [79]. The evolutionary conservation of these structural and functional motifs highlights the key role of CCR5 in immune surveillance, leukocyte chemotaxis, and host defense, as variations in these sequences often lead to reduced chemokine binding, disrupted signaling cascades, and increased susceptibility to infectious and inflammatory diseases [80,81]. Nevertheless, genetic variants influence its activity. The most prominent example is the CCR5-Δ32 deletion, a 32-base pair frameshift mutation that results in a truncated, nonfunctional receptor that fails to reach the cell surface [82,83]. Homozygosity for this mutation confers near-complete resistance to HIV-1 infection, as the retrovirus necessitates the CCR5 as a major co-receptor for entry into CD4+ T cells and macrophages [84]. Heterozygous individuals exhibit delayed disease progression, reflecting a dose-dependent relationship between CCR5 surface density and viral infectivity [85]. In addition to HIV-1 infection, CCR5 polymorphisms influence susceptibility to other infectious diseases, such as West Nile, SARS-CoV-2, and HBV viruses [86,87,88], as well as in inflammatory and autoimmune disorders characterized by dysregulated chemokine signaling [89,90].

2.2. Distribution of the Chemokine CCR5 Receptor

The CCR5 receptor displays a complex and finely tuned distribution pattern across immune and non-immune systems, reflecting its central role in mediating leukocyte trafficking [91], shaping inflammatory microenvironments [92], and influencing host–pathogen interactions [93]. Its expression is neither static nor homogeneous; rather, it is dynamically regulated by cellular activation states, tissue context, cytokine milieu, and pathological conditions [94].
Within the immune system, CCR5 expression is most prominently localized to activated and memory T lymphocytes, with a particularly high prevalence among CD4+ T cell subsets exhibiting a Th1-polarized functional phenotype [95,96]. CCR5+ CD4+ T cells exhibit a significant capacity for directed migration toward sites of antigen presentation and microbial insult, a process orchestrated by chemotactic gradients of the high-affinity ligands (e.g., CCL3, CCL4, and CCL5) [97]. Moreover, several subsets of CD8+ T lymphocytes express CCR5, and their receptor-dependent chemotactic responsiveness allows the targeted mobilization of cytotoxic effector functions within virally infected or inflamed tissue microenvironments [98]. This dual presence in both helper and cytotoxic T cells ensures that CCR5 orchestrates coordinated adaptive immune responses, tightly linking antigen recognition with tissue-directed homing [99].
Monocytes and macrophages further exemplify the functional importance of CCR5 distribution. Circulating monocytes, particularly the inflammatory CD14+CD16 subset, upregulate CCR5 upon activation and during differentiation into macrophages, enabling their recruitment into tissues where inflammatory mediators are present [100]. Tissue-resident macrophages, such as those in the gut lamina propria [101], the liver [102], and the lung [103], also express CCR5 under pro-inflammatory conditions, enhancing local immune surveillance and contributing to persistent inflammatory signaling pathways. Dendritic cells (DCs), especially immature subsets populating epithelial surfaces, utilize CCR5 not only to migrate into lymphoid structures but also to associate with other immune cells within inflamed tissues [104]. Similarly, NK cells use CCR5 to migrate toward virally infected or neoplastic tissues, thus facilitating the execution of innate immune effector functions [105].
Although historically considered an immune-cell-restricted receptor, CCR5 has been observed in several non-hematopoietic compartments under pathological circumstances. Endothelial cells might express CCR5 during vascular inflammation [106], where receptor activation promotes leukocyte adhesion and transmigration across the endothelium [107]. Similarly, epithelial cells in inflamed mucosal surfaces can transiently upregulate CCR5, possibly serving as amplifiers of immune signaling [108]. In the CNS, CCR5 expression has been distinguished in microglia [109], astrocytes [43], and infiltrating leukocytes during neuroinflammatory events [110], where its presence is linked to glial activation, neuronal damage, and compromise of BBB integrity.
Even within cancer biology, CCR5 has been implicated in a wide spectrum of malignancies, including breast [111], prostate [112], colorectal [113], pancreatic [114], and gastric cancers [115], as well as hematological malignancies such as acute myeloid leukemia [116] and multiple myeloma [117]. Its expression has been linked to tumor-promoting inflammation, metastatic dissemination, and modulation of the tumor microenvironment, suggesting a broad relevance across both solid tumors and hematologic neoplasms [118].

2.3. Functional Significance of CCR5 Receptor

2.3.1. Role in Chemotaxis and Immune Cell Trafficking

CCR5-driven chemotaxis plays a key role in shaping adaptive immune responses by directing the migration of CD4+ and CD8+ T lymphocytes, monocytes, and DCs to sites of inflammation or infection [119]. This redistribution of immune cells ensures that effector populations are concentrated in peripheral tissues where microbial antigens, danger signals, or tissue damage are present [120]. Through directing the activity of Th1 and Th17 subsets, CCR5 signaling promotes cell-mediated immunity, the production of pro-inflammatory cytokines (e.g., IFN-γ, TNF-α, and IL-17), and the clearance of intracellular pathogens [121,122]. In monocytes and DCs, CCR5 activation allows tissue infiltration, where these cells undergo differentiation and antigen presentation, thereby bridging innate and adaptive immune compartments [104,123].
Beyond its role in cell trafficking, CCR5 supports the structural and functional stabilization of immunological synapses between T cells and antigen-presenting cells (APCs) [124]. Mechanistically, CCR5 signaling enhances the affinity of integrins (e.g., integrin α4), thereby strengthening cell–cell adhesion [125]. This supports prolonged T cell-APC interactions, efficient antigen recognition, and sustained TCR signaling, which are crucial for effective clonal expansion and effector differentiation [126]. CCR5-orchestrated cytoskeletal remodeling further reinforces the polarization of signaling complexes and the directional secretion of cytokines and lytic granules toward target cells [127].
In the CNS, CCR5 expression on resident microglia, perivascular macrophages, and infiltrating monocytes regulates leukocyte recruitment during neuroinflammatory conditions [42,43,128]. Under physiological conditions, this contributes to the clearance of pathogens and debris, as well as the resolution of acute insults (e.g., viral encephalitis or traumatic injury) [129]. However, dysregulated CCR5 activation has deleterious effects. Persistent chemokine gradients drive continuous immune cell infiltration, excessive release of ROS, NO, and pro-inflammatory cytokines, ultimately amplifying neuroinflammation and contributing to neuronal dysfunction and loss [130].

2.3.2. CCR5 as a Co-Receptor for HIV-1 Entry

One of the most clinically significant functions of CCR5 lies in its role as a principal co-receptor for HIV-1 [131]. Initially, the HIV-1 envelope glycoprotein gp120 binds to the CD4 receptor on CD4+ T cells, macrophages, and DCs. This binding promotes conformational rearrangements in gp120, especially in the V1/V2 region, which exposes the V3 loop. The V3 loop subsequently interacts with the extracellular domains and transmembrane regions of CCR5, mainly the N-terminal sulfated tyrosines and the ECL2 [132,133].
The interaction between gp120 and CCR5 induces further conformational rearrangements that expose the gp41 fusion peptide. Following this, gp41 embeds its fusion domain into the target cell membrane and adopts a six-helix bundle conformation through extensive structural refolding [134]. This conformational change juxtaposes the viral and target cell membranes, driving membrane fusion and the creation of a fusion pore. Through this pore, the viral capsid is delivered into the cytoplasm, thus initiating the HIV-1 replication cycle [135].

2.3.3. CCR5 in Inflammation and Autoimmunity

Beyond its classical role in mediating chemotactic responses, CCR5 is involved in the regulation and fine-tuning of inflammatory processes [39]. Dysregulated CCR5 signaling can precipitate maladaptive immune responses, thereby contributing to the pathogenesis of autoimmune disorders, such as rheumatoid arthritis (RA), multiple sclerosis (MS), and systemic lupus erythematosus (SLE). In these pathologies, CCR5-expressing immune cells perpetuate tissue injury through sustained infiltration, local production of pro-inflammatory cytokines and ROS, collectively amplifying inflammation and promoting chronic tissue damage [136,137,138].

2.3.4. CCR5 in Tissue Repair and Regeneration

Emerging evidence indicates that CCR5 plays a multifaceted role in tissue repair and regeneration, extending beyond its well-characterized functions in immune cell trafficking and inflammatory modulation [106,139]. During the reparative response, CCR5 mediates the directed migration of various reparative cell populations, including macrophages and mesenchymal stem cells (MSCs), to regions of injured tissue. This recruitment is mediated by CCR5 binding to its ligands, which establish chemotactic gradients within the damaged tissue microenvironment [140]. Following recruitment, CCR5-expressing macrophages promote the resolution of inflammation by undergoing polarization from a pro-inflammatory M1 phenotype to an M2 reparative/anti-inflammatory phenotype [141]. M2 macrophages secrete anti-inflammatory cytokines, such as IL-10 and TGF-β, and produce MMPs, thus facilitating remodeling of the ECM [142]. In parallel, CCR5 activation promotes angiogenesis via the production of VEGF, thereby promoting restoration of tissue perfusion and structural integrity [139].

2.3.5. CCR5 in Stem Cell and Hematopoietic Regulation

CCR5 has been increasingly recognized as an important modulator of hematopoietic stem and progenitor cell (HSPC) dynamics [143]. Beyond its well-established role in leukocyte trafficking and immune cell activation, CCR5 contributes to the regulation of migration, homing, and retention of HSPCs within the bone marrow niche [144]. HSPCs reside in specialized microenvironments where interactions with stromal cells, extracellular matrix components, and soluble factors are critical for maintaining stemness, quiescence, and differentiation potential [145]. CCR5 activation shapes these processes by modulating chemotactic signaling and driving cytoskeletal rearrangements that support the directed migration and spatial organization of cells within the niche [146].
Under homeostatic conditions, CCR5 appears to have a relatively modest role compared to other chemokine receptors (e.g., CXCR4). However, its functional importance is markedly amplified under inflammatory conditions [147]. In the context of infection, tissue injury, or subsequent to myeloablative conditioning regimens, CCR5 expression in the HSPC compartment can be upregulated, enhancing their responsiveness to inflammatory chemokines and promoting mobilization from or recruitment back to the bone marrow [148].
Furthermore, CCR5-mediated signaling has been implicated in the crosstalk between HSPCs and the immune system. Pro-inflammatory cytokines can induce CCR5 expression on HSPCs, which in turn allows these cells to sense and migrate toward inflamed tissues [149].

3. The Role of CCR5 in the Pathophysiology of Neuropathic Pain

3.1. CCR5-Mediated Peripheral Mechanisms in Neuropathic Pain

Peripheral nerve injury constitutes a critical event that initiates a complex cascade of cellular and molecular responses within the PNS, which collectively contribute to the initiation and persistence of neuropathic pain [150]. The initial mechanical or chemical insult results in axonal degeneration, Wallerian degeneration, and demyelination, accompanied by the release of DAMPs, ATP, and pro-inflammatory cytokines from both neuronal and non-neuronal cells [151]. These factors create a highly chemotactic environment that promotes the recruitment and activation of several immune cells, including monocytes, macrophages, neutrophils, and T lymphocytes [152].
Concurrently, CCR5 expression is upregulated on both infiltrating immune cells and resident sensory neurons within the dorsal root ganglia (DRG). The engagement of CCR5 on immune cells not only governs chemotaxis but also triggers intracellular signaling cascades that result in the transcriptional and translational upregulation of numerous inflammatory mediators, such as pro-inflammatory cytokines, prostaglandins, leukotrienes, and ROS [153,154,155]. Collectively, these mediators establish an inflammatory milieu (known as inflammatory soup) that potentiates nociceptor sensitization and contributes to peripheral hyperexcitability [156].
On the other hand, CCR5 exerts multifaceted effects on sensory neurons, modulating their immediate excitability and long-term functional plasticity. Although not yet entirely established, CCR5-mediated activation of nociceptors is likely to promote the activation of VGCCs, VGSCs, and TRPs. VGSCs (e.g., Nav1.7, Nav1.8, and Nav1.9) experience transcriptional upregulation and post-translational phosphorylation, strengthening their conductance and lowering the threshold for action potential generation [157]. VGCCs (e.g., Cav2.2 and Cav3.2) are equivalently modulated, increasing intracellular Ca2+ influx, which potentiates presynaptic release of neurotransmitters and potentiates abnormal excitability [158]. TRP channels (e.g., TRPV1, TRPA1, and TRPM8) are sensitized via direct phosphorylation by MAPKs or PKA/PKC signaling, enhancing responsiveness to thermal, mechanical, and chemical stimuli [159]. Ultimately, nociceptor activation results in the release of excitatory neurotransmitters onto the spinal cord, driving central sensitization and pain transmission. Primary afferent terminals within the dorsal horn discharge glutamate, substance P, and CGRP, which act on postsynaptic neurons and surrounding glial cells [160]. Moreover, substance P promotes peripheral immune cell recruitment and local inflammation, leading to decreased excitation thresholds in non-nociceptive fibers. This process facilitates aberrant neuronal activation and contributes to peripheral sensitization, thereby amplifying pain perception and driving hyperalgesia and allodynia [161].
Glial cells, including satellite glial cells and Schwann cells in injured nerve fibers, also express CCR5 and are critical mediators of peripheral sensitization following nerve injury [55,162]. Based on investigations conducted with other chemokine receptors (e.g., CCR2 and CX3CR1), CCR5 signaling in these glial cells probably mobilizes intracellular Ca2+ and activates PLC-γ and PI3K-Akt pathways, thereby promoting the release of pro-inflammatory cytokines (e.g., TNF-α and IL-1β) and pro-inflammatory mediators (e.g., prostaglandin E2) [163,164]. TNF-α and IL-1β can upregulate the membrane trafficking and surface expression of VGSCs, particularly Nav1.7 and Nav1.8, in DRG neurons [165,166]. Mechanistically, TNF-α binds to TNFR1/2 receptors, leading to activation of downstream signaling cascades such as the p38 MAPK and ERK1/2 pathways [167]. These kinases phosphorylate specific Ser/Thr residues on Nav1.7 and Nav1.8, increasing channel open probability and accelerating recovery from inactivation [168]. IL-1β, through IL-1RI, activates MyD88-dependent signaling, resulting in the activation of IRAK1/4, TRAF6, and NF-κB, thus promoting the transcriptional upregulation of VGSCs [169]. IL-6 and PGE2 modulate several GPCRs expressed in DRG neurons, with IL-6 signaling mediated via its receptor complex (IL-6Rα/gp130) and PGE2 signaling through EP2 and EP4 receptors [170,171]. Activation of these receptors stimulates adenylate cyclase, increasing intracellular cAMP levels and activating PKA. PKA phosphorylates TRP channels, like TRPV1, on cytoplasmic Ser/Thr residues, reducing their activation threshold and promoting channel opening in response to thermal stimuli. Concurrently, Gq/11-coupled EP receptors activate PLC, leading to the generation of DAG and IP3, which subsequently activate PKC and mobilize intracellular Ca2+ stores. PKC-mediated phosphorylation further sensitizes TRPV1 and other TRP channels, thus enhancing Ca2+ influx and contributing to thermal hyperalgesia [172]. NO, produced by inducible nitric oxide synthase (iNOS) in glial cells, together with ROS generated via NADPH oxidase or mitochondrial pathways, modulate the gating properties of multiple ion channels in DRG neurons [173]. NO activates guanylyl cyclase, increasing cGMP levels and stimulating protein kinase G (PKG), which phosphorylates and modifies the activity of VGSCs and K+ channels, thus enhancing excitatory currents [174]. Additionally, NO and ROS enhance synaptic glutamate release, further potentiating nociceptive signaling [175,176].
Therefore, CCR5 represents a likely key node in peripheral neuropathic pain mechanisms by integrating chemokine-mediated immune recruitment, glial-neuronal crosstalk, intracellular kinase signaling, ion channel modulation, and transcriptional upregulation of pronociceptive mediators. All of these events establish and maintain a hyperexcitable peripheral nociceptive network that supports the persistence and chronicity of neuropathic pain, making CCR5 a promising target for therapeutic interventions aimed at disrupting maladaptive peripheral sensitization.

3.2. CCR5-Mediated Central Mechanisms in Neuropathic Pain

Following peripheral sensitization, nociceptive inputs originating from damaged tissues are transmitted via primary afferent fibers to the dorsal horn of the spinal cord. Upon arrival, these signals initiate various molecular and cellular responses that modulate pain processing [177]. Notably, CCR5 expression is markedly elevated in both dorsal horn neurons and resident microglial populations. This upregulation is temporally correlated with the progression of central sensitization, suggesting a critical modulatory role for CCR5 in the amplification and maintenance of nociceptive signaling [54,178].
Within the spinal dorsal horn, CCR5 contributes to central sensitization through multiple convergent mechanisms that involve intricate neuroimmune and neuroglial interactions (Figure 2): (1) microglial activation: induction of CCR5 (by CCL3, CCL4, and CCL5) activates intracellular signaling cascades, like the Gi/o protein-dependent inhibition of adenylate cyclase, resulting in decreased cAMP levels, and the downstream modulation of PI3K/Akt and ERK1/2 MAPK pathways [42,179]. All of these processes promote microglial proliferation and phenotypic activation, marked by increased expression of Iba1, CD11b, and MHC-II [180]. Activated microglia release a repertoire of pro-inflammatory mediators that recruit additional immune cells into the spinal cord parenchyma [181]. Importantly, CCR5-driven signaling enhances the release of BDNF, which acts on neuronal TrkB receptors. BDNF-TrkB pathway activation downregulates the neuronal KCC2 cotransporter in dorsal horn projection neurons, resulting in a depolarizing shift in the Cl reversal potential and thus inhibiting GABAergic and glycinergic transmission [182,183,184]. This dysregulation of the excitatory-inhibitory balance constitutes a key mechanism of central sensitization [185]; (2) synaptic plasticity: CCR5-mediated signaling in dorsal horn neurons regulates activity-dependent synaptic plasticity by modulating the function and trafficking of glutamatergic receptor systems [186]. Upon ligand binding, CCR5 signaling induces pERK, which facilitates NMDA receptor complex activation [187,188]. This fact strengthens NMDA receptor channel conductance and extends channel open time, thus facilitating Ca2+ influx [189]. Increased intracellular Ca2+ activates signaling molecules like CaMKIV and CREB, which regulate transcription of plasticity-related genes (e.g., c-fos, Arc, and BDNF) [190]. Simultaneously, CCR5 engagement drives the trafficking of GluA1-containing AMPA receptors to the postsynaptic membrane. These molecular modifications consolidate a state of LTP-like plasticity in nociceptive pathways of the dorsal horn [191,192]; (3) neuron-glia feedback loops: neurons within the dorsal horn that express CCR5 respond to chemokine ligands by activating NF-κB and AP-1 transcriptional programs, leading to de novo production of CCL2, CCL5, and other pro-inflammatory mediators [186,193,194]. These chemokines function via autocrine and paracrine mechanisms, thereby further activating CCR5 on adjacent microglia and astrocytes [195]. Moreover, CCR5 signaling modulates presynaptic release machinery through Gβγ subunit interactions with VGCCs, facilitating neurotransmitter release [186]. This fact enhances excitatory drive onto microglia and astrocytes, which respond with further cytokine and chemokine secretion [186,196]. Although the exact relationship remains unclear, astrocytic CCR5 activation likely promotes glutamate release through the system Xc antiporter and connexin hemichannels, thus intensifying excitotoxic signaling in dorsal horn neurons. This neuroimmune cross-talk establishes a self-reinforcing feedback loop, which is fundamental for the transition from acute nociceptive signaling to chronic neuropathic pain, a state characterized by chronic neuroinflammation and maladaptive synaptic plasticity.
Finally, CCR5 signaling might play a crucial role in supraspinal pain modulation. Current evidence indicates that CCR5 signaling may modulate descending pain control pathways projecting from the periaqueductal gray (PAG), an essential brainstem structure regulating intrinsic pain pathways [197]. Within the PAG, CCR5 expression may influence the balance between excitatory and inhibitory neurotransmission, due to the inhibitory effect exerted on μ-opioid receptors [56].

3.3. Interplay Between CCR5 and Other Chemokine Receptors in the Modulation of Neuropathic Pain

CCR5 serves as a central integrator within the chemokine receptor network, orchestrating glial activation, neuronal sensitization, and immune cell trafficking to regulate the onset and development of neuropathic pain [39,42,43,104,106,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149]. Its crosstalk with other chemokine receptors establishes an integrated regulatory system that potentiates microglial and astrocytic responses while leukocyte recruitment to sites of neuronal injury [198,199]. This network regulates pro-inflammatory signaling yet may facilitate maladaptive neuroplasticity when disrupted.
CCR2, the primary receptor for CCL2, is integrated with CCR5 signaling, given their co-expression on spinal microglia, astrocytes, DRG neurons, and infiltrating monocytes, where their concomitant activation induces synergistic intracellular pathways [200,201]. CCR5 engagement primes CCR2-dependent pathways such as G-protein-mediated Ca2+ influx, MAPK/ERK phosphorylation, and NF-κB nuclear translocation, leading to enhanced production of pro-inflammatory cytokines (such as IL-1β, IL-6, and TNF-α), chemokines (e.g., CCL3 and CCL5), while CCR2 activation reciprocally regulates CCR5 surface expression and ligand sensitivity, orchestrating a feed-forward loop that sustains microglial reactivity and central sensitization [202]. Indeed, some studies support the crosstalk between CCR2 and CCR5. In CCR5-lacking mice, CCR5 deficiency promotes LPS-induced astrogliosis, Aβ plaque deposition, and memory impairment due to increased CCR2 expression in the CNS [203]. Moreover, the involvement of CCR2 and CCR5 has been shown in preclinical models of obesity-induced hypersensitivity and diabetic neuropathy, where administration of dual CCR2/CCR5 antagonists led to pain reduction [204].
Beyond CCR2, CCR5 also shows functional crosstalk with CXCR3, a receptor for the IFN-inducible chemokines CXCL9, CXCL10, and CXCL11, which mediates T-cell recruitment to the spinal cord and DRG [205]. CCR5 influences CXCR3 signaling indirectly by modulating the local chemokine milieu, upregulating CXCR3 ligand expression in resident glia and infiltrating lymphocytes, and potentially co-localizing with CXCR3 on microglia, thereby coordinating downstream signaling several cascades including PI3K/Akt, ERK1/2, and JAK/STAT pathways that enhance glial activation, sustain pro-inflammatory cytokine release, and facilitate the persistence of neuropathic hyperalgesia [206].
On the other hand, CCR5 displays functional redundancy with CCR1 and CCR3, supporting overlapping ligand binding, mainly with CCL5 and CCL3, which can compensate for CCR5 blockade and maintain chemokine-mediated microglial activation and leukocyte recruitment [186]. This redundancy underscores the chemokine signaling network and accounts for the partial analgesic effects seen with CCR5 antagonists.
CCR5 interacts with other chemokine receptors to regulate inflammatory signaling, but dysregulation of this network can induce maladaptive neuroplasticity when it becomes dysregulated, driving chronic neuropathic pain marked by allodynia, hyperalgesia, and spontaneous pain [207]. Multi-target strategies concurrently inhibiting CCR5 and CCR2, CXCR3, or CX3CR1 suppress compensatory signaling, attenuate glial activation, and control maladaptive neuroinflammation without inducing immune suppression.

4. CCR5 Inhibition: Emerging Approaches for Analgesia

4.1. Evidence from Animal Models of Neuropathic Pain

Emerging evidence increasingly implicates CCR5 as a critical regulator of neuroinflammatory pathways that contribute to the initiation and maintenance of neuropathic pain. Preclinical studies using diverse rodent models of neuropathic pain, like CCI, PSNL, CIPN, and DPN, have shown that modulation of CCR5 attenuates pathological pain behaviors. Genetic deletion of CCR5, pharmacological antagonism with small molecules or monoclonal antibodies, and targeted knockdown via RNA interference all result in significant reductions in mechanical allodynia, thermal hyperalgesia, and spontaneous pain behaviors. The following table provides a comprehensive overview of recent preclinical investigations evaluating CCR5-targeted interventions across diverse neuropathic pain models.
Table 1 shows an overview of the representative CCR2/CCR5-targeting compounds, while Table 2 summarizes their pharmacological characteristics, clinical indications, mechanisms of action, pharmacokinetic profiles, and clinical development status, and Table 3 presents the adverse effects reported following their administration.

4.2. Translational Potential and Early Clinical Findings

Clinical evidence for CCR5 blockade in neuropathic pain remains at an early stage. Maraviroc has received FDA approval for long-term use in HIV-1 infection [215] (Table 2), with a generally acceptable safety profile that includes routine hepatotoxicity monitoring and careful consideration of concomitant medications due to CYP3A4-mediated drug–drug interactions [216]. However, several pharmacological and clinical limitations restrict its suitability for chronic pain indications. Idiosyncratic hepatotoxicity, although uncommon, has been documented and necessitates ongoing hepatic function surveillance during long-term treatment (Table 3) [226]. Moreover, maraviroc displays relatively low oral bioavailability, largely attributable to extensive first-pass hepatic metabolism, which complicates dose optimization and contributes to interindividual variability in systemic exposure [229]. An additional limitation is its reduced permeability across the BBB. Although maraviroc exerts immunomodulatory effects in peripheral tissues, its constrained CNS penetration raises concerns regarding adequate engagement of CCR5 within spinal and supraspinal regions that mediate pain processing, where CCR5-driven neuroinflammatory cascades are central to the onset and development of neuropathic pain [230]. Consequently, attaining therapeutically meaningful CCR5 inhibition in the CNS with maraviroc might require elevated systemic exposure, thus increasing the risk of hepatotoxicity and unfavorable pharmacokinetic interactions.
These limitations have driven intensified investigation into alternative CCR5 blockade strategies. Monoclonal antibodies targeting CCR5 have emerged as promising candidates due to high target specificity, reduced hepatic metabolism, and extended systemic half-life, potentially enabling more consistent receptor occupancy. Unlike small-molecule antagonists, specific monoclonal antibodies exhibit increased pharmacokinetic properties and, when optimally engineered, may enable more efficient modulation of neuroimmune signaling [231]. While randomized controlled trials of maraviroc in neuropathic pain have yet to be conducted, its thoroughly defined clinical pharmacology profile supports assessment in proof-of-concept studies and reinforces the potential of next-generation CCR5 inhibitors, like monoclonal antibodies, as promising therapeutic candidates for chronic pain.
Monoclonal antibodies directed against CCR5, including leronlimab (PRO-140), have been tested in clinical populations like HIV-1, oncology, and COVID-19 [232,233,234]. These studies confirm sustained receptor occupancy, predictable pharmacokinetics, and a favorable safety profile. Although no neuropathic pain-specific clinical data are currently available, the immunomodulatory actions of leronlimab align well with mechanistic pathways implicated in neuroinflammatory pain. The long-lasting therapeutic effect and receptor interaction of monoclonal antibodies could be helpful in chronic pain diseases requiring suppression of immune-neuronal signaling [235].

5. Future Perspectives

The evolving understanding of CCR5’s role in neuropathic pain opens multiple avenues for future research and therapeutic development. A key focus is the identification of predictive biomarkers to enable precise patient stratification [236]. By elucidating molecular signatures, chemokine profiles, or genetic polymorphisms associated with enhanced CCR5 signaling, researchers may be able to determine which patient populations are most likely to benefit from CCR5-targeted interventions. This precision medicine approach has the potential to optimize clinical trial design and improve therapeutic outcomes [237].
Another promising avenue is the investigation of combinatorial strategies with existing analgesics. Integrating CCR5 antagonists with conventional pharmacotherapies (such as opioids) might produce synergistic effects and enhance analgesic efficacy while reducing doses and minimizing adverse events [238]. Systematic preclinical and translational studies are required to identify optimal combinations, dosing regimens, and timing of administration to maximize therapeutic benefit and minimize toxicity.
The development of next-generation CCR5 modulators is equally important. Current pharmacological agents, while effective, may have limitations in selectivity, pharmacokinetics, or tissue penetration [239]. Rational drug design approaches, including structure-based improvement [240], allosteric modulators [241], and monoclonal antibodies [242], could yield compounds with enhanced receptor specificity, improved CNS penetration, and prolonged half-life, thereby improving clinical efficacy and safety profiles.
Moreover, advanced translational models should be employed to bridge the gap between preclinical findings and clinical application. Humanized mouse models [243], patient-derived organoids [244], and high-throughput screening platforms [245] could provide insights into CCR5 biology in human tissues, helping to refine therapeutic strategies. Additionally, longitudinal clinical studies will be crucial to evaluate long-term safety, efficacy, and potential off-target effects of CCR5-targeted therapies [208].
Finally, a multidisciplinary approach integrating immunology, neuroscience, pharmacology, and clinical pain research will be fundamental for accelerating the translation of CCR5 antagonists from bench to bedside [246]. Through these concerted efforts, CCR5-targeted therapies hold the potential not only to ameliorate neuropathic pain but also to redefine mechanistically informed treatment paradigms for patient’s refractory to conventional analgesics.

6. Conclusions

CCR5 has emerged as a critical mediator of neuroinflammatory processes underlying the development and maintenance of neuropathic pain. Preclinical evidence from diverse rodent models demonstrates that CCR5 contributes to central sensitization through activation of glial cells and several neuronal populations, as well as peripheral sensitization through recruitment of immune cells to injured nerves and modulation of DRG neurons. Pharmacological antagonism, genetic deletion, or targeted knockdown of CCR5 alleviates mechanical allodynia, thermal hyperalgesia, and spontaneous pain behaviors, highlighting its pivotal role in pain pathophysiology.
These findings are encouraging and justify continued investigation of CCR5 signaling in nociceptive modulation; however, major gaps in mechanistic insight and translational feasibility persist. Some referenced preclinical studies present methodological weaknesses that limit interpretability and external validity. Common limitations include small sample sizes, huge reliance on rodent models without validation in higher-order species, and inadequate behavioral controls or standardized pain assessment protocols. As a result, reproducibility and internal validity are uncertain, and extrapolation to human pain physiology remains limited.
To address these issues, future research must implement more rigorous experimental designs, including appropriate statistical power, randomization, blinding, and validated behavioral assays. Translational studies are needed to define dosing strategies, pharmacokinetic and pharmacodynamic profiles, and long-term safety, particularly regarding the immunomodulatory risks of chronic CCR5 inhibition. Ultimately, randomized controlled trials in humans will be essential to establish therapeutic efficacy across diverse pain conditions, including neuropathic, inflammatory, and cancer-related pain.
CCR5 antagonists hold therapeutic value by targeting neuroimmune pathways and introducing a novel intervention strategy for neuropathic pain syndromes that do not respond to conventional analgesic therapies. Compounds including maraviroc and experimental monoclonal antibodies have demonstrated efficacy in preclinical models, providing a strong rationale for clinical translation. Given the multifactorial nature of chronic pain, future investigations should also evaluate combinatorial or synergistic approaches that integrate CCR5-targeted therapies with standard analgesics (e.g., opioids, gabapentinoids, or anti-inflammatory agents) to enhance efficacy while minimizing adverse effects and tolerance.
In summary, CCR5 represents a promising molecular target for next-generation analgesics; however, clinical implementation will require methodologically rigorous, interdisciplinary research that bridges preclinical findings with patient-centered outcomes.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
[Cli]Intracellular Cl concentration
7TMSeven transmembrane
AktProtein kinase B
ALTAlanine aminotransferase
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
AP-1Activator protein 1
APCAntigen-presenting cell
ArcActivity-regulated cytoskeleton-associated protein
ASTAspartate aminotransferase
ATPAdenosine triphosphate
BBBBlood–brain barrier
BDNFBrain-derived neurotrophic factor
c-fosCellular oncogene fos
Ca2+Calcium ion
CaMKCalcium/calmodulin-dependent protein
CaMKIVCalcium/calmodulin-dependent protein kinase IV
cAMPCyclic adenosine monophosphate
cGMPCyclic guanosine monophosphate
Cav2.2Voltage-gated calcium channel subtype 2.2
CCIChronic constriction injury
CCL12Chemokine (C-C motif) ligand 12
CCL2Chemokine (C-C motif) ligand 2
CCL3Chemokine (C-C motif) ligand 3
CCL4Chemokine (C-C motif) ligand 4
CCL5Chemokine (C-C motif) ligand 5
CCL7Chemokine (C-C motif) ligand 7
CCL8Chemokine (C-C motif) ligand 8
CCR1C-C chemokine receptor type 1
CCR2C-C chemokine receptor type 2
CCR3C-C chemokine receptor type 3
CCR5C-C chemokine receptor type 5
CD11bCluster of differentiation 11b
CD14Cluster of differentiation 14
CD16Cluster of differentiation 16
CD4Cluster of differentiation 4
CD8Cluster of differentiation 8
CGRPCalcitonin gene-related peptide
ClChloride ion
CNSCentral nervous system
CREBcAMP response element-binding protein
CXCL9C-X-C motif chemokine ligand 9
CXCL10C-X-C motif chemokine ligand 10
CXCL11C-X-C motif chemokine ligand 11
CXCR3C-X-C chemokine receptor type 3
CX3CR1C-X3-C motif chemokine receptor 1
DAGDiacylglycerol
DAMPDamage-associated molecular pattern
DAPTAD-Ala1-peptide T-amide
DCDendritic cell
DPNDiabetic peripheral neuropathy
DRGDorsal root ganglia
ECL1Extracellular loop 1
ECL2Extracellular loop 2
ECL3Extracellular loop 3
ECMExtracellular matrix
EP2Prostaglandin E2 receptor subtype 2
EP4Prostaglandin E2 receptor subtype 4
ERKExtracellular signal-regulated kinase
ERK1/2Extracellular signal-regulated kinases 1 and 2
FDAFood and Drug Administration
GABAGamma-aminobutyric acid
GFAPGlial fibrillary acidic protein
GluA1Glutamate receptor subunit A1 (AMPA receptor subunit)
gp120Glycoprotein 120
G0p130Glycoprotein 130
gp41Glycoprotein 41
GPCRG protein-coupled receptor
GRKG protein-coupled receptor kinase
GαiG protein alpha i subunit
GαqG protein alpha q subunit
GβγG protein beta-gamma subunits
Gq/11G protein subtypes q and 11
HBVHepatitis B virus
HIV-1Human immunodeficiency virus type 1
i.t.Intrathecal injection
IASPInternational Association for the Study of Pain
Iba1Ionized calcium-binding adapter molecule 1
ICAM-1Intercellular adhesion molecule 1
ICL1Intracellular loop 1
ICL2Intracellular loop 2
ICL3Intracellular loop 3
IFN-γInterferon gamma
IL-1RIInterleukin-1 receptor type I
IL-10Interleukin 10
IL-17Interleukin 17
IL-18Interleukin 18
IL-18BPInterleukin 18 binding protein
IL-1βInterleukin 1 beta
IL-1RAInterleukin 1 receptor antagonist
IL-6Interleukin 6
IL-6RαInterleukin-6 receptor alpha
iNOSInducible nitric oxide synthase
IP3Inositol 1,4,5-trisphosphate
IRAK1/4Interleukin-1 receptor-associated kinases 1 and 4
JAKJanus kinase
K+Potassium ion
KCCPotassium chloride cotransporter
KCC2Potassium chloride cotransporter 2
LTPLong-term potentiation
M1Pro-inflammatory microglial phenotype
M2Anti-inflammatory microglial phenotype
MAPKMitogen-activated protein kinase
MASHMetabolic dysfunction-associated steatohepatitis
MHC-IIMajor histocompatibility complex class II
MIP-1αMacrophage inflammatory protein 1 alpha
MIP-1βMacrophage inflammatory protein 1 beta
MMPMatrix metalloproteinase
MSMultiple sclerosis
MSCMesenchymal stem cell
MyD88Myeloid differentiation primary response 88
mTORMammalian target of rapamycin
Nav1.3Voltage-gated sodium channel subtype 1.3
Nav1.7Voltage-gated sodium channel subtype 1.7
Nav1.8Voltage-gated sodium channel subtype 1.8
NADPHNicotinamide adenine dinucleotide phosphate
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NGFNerve growth factor
NKNatural killer
NMDAN-methyl-D-aspartate
NONitric oxide
NOS2Nitric oxide synthase 2
p38 MAPKp38 mitogen-activated protein kinase
pERKPhosphorylated extracellular signal-regulated kinase
PGE2Prostaglandin E2
PI3KPhosphoinositide 3-kinase
PI3K-βPhosphatidylinositol 3-kinase beta
PI3K-γPhosphatidylinositol 3-kinase gamma
PIP2Phosphatidylinositol 4,5-bisphosphate
PIPNPaclitaxel-induced peripheral neuropathy
PKPharmacokinetics
PKAProtein kinase A
PKBProtein kinase B
PKCProtein kinase C
PKGProtein kinase G
PLC-βPhospholipase C beta
PLC-γPhospholipase C gamma
PNSPeripheral nervous system
PRRPattern recognition receptor
PSNLPartial sciatic nerve ligation
qRT-PCRQuantitative real-time Polymerase Chain Reaction
RARheumatoid arthritis
RANTESRegulated upon activation, normal T cell expressed and secreted
ROSReactive oxygen species
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SerSerine
SLESystemic lupus erythematosus
STATSignal transducer and activator of transcription
STAT3Signal transducer and activator of transcription 3
STZStreptozotocin
TCRT cell receptor
TGF-βTransforming growth factor beta
Th1T helper 1 cell
Th17T helper 17 cell
ThrThreonine
TM3Transmembrane helix 3
TM5Transmembrane helix 5
TM6Transmembrane helix 6
TNF-αTumor necrosis factor alpha
TNFR1Tumor necrosis factor receptor 1
TNFR2Tumor necrosis factor receptor 2
TRAF6TNF receptor-associated factor 6
TrkBTropomyosin receptor kinase B
TRPTransient receptor potential channel
TRPA1Transient receptor potential ankyrin 1
TRPM8Transient receptor potential melastatin 8
TRPV1Transient receptor potential vanilloid 1
V1Variable region 1 of gp120
V2Variable region 2 of gp120
V3Variable region 3 of gp120
VEGFVascular endothelial growth factor
VGCCVoltage-gated calcium channel
VGSCVoltage-gated sodium channel
XcCystine/glutamate antiporter

References

  1. Raja, S.N.; Carr, D.B.; Cohen, M.; Finnerup, N.B.; Flor, H.; Gibson, S.; Keefe, F.J.; Mogil, J.S.; Ringkamp, M.; Sluka, K.A.; et al. The revised International Association for the Study of Pain definition of pain: Concepts, challenges, and compromises. Pain 2020, 161, 1976–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Vader, K.; Bostick, G.P.; Carlesso, L.C.; Hunter, J.; Mesaroli, G.; Perreault, K.; Tousignant-Laflamme, Y.; Tupper, S.; Walton, D.M.; Wideman, T.H.; et al. The Revised IASP Definition of Pain and Accompanying Notes: Considerations for the Physiotherapy Profession. Physiother. Can. 2021, 73, 103–106. [Google Scholar] [CrossRef] [PubMed]
  3. Fashler, S.R.; Cooper, L.K.; Oosenbrug, E.D.; Burns, L.C.; Razavi, S.; Goldberg, L.; Katz, J. Systematic Review of Multidisciplinary Chronic Pain Treatment Facilities. Pain Res. Manag. 2016, 2016, 5960987. [Google Scholar] [CrossRef] [PubMed]
  4. Bonezzi, C.; Fornasari, D.; Cricelli, C.; Magni, A.; Ventriglia, G. Not All Pain is Created Equal: Basic Definitions and Diagnostic Work-Up. Pain Ther. 2020, 9, 1–15. [Google Scholar] [CrossRef]
  5. Sima, S.; Lapkin, S.; Gan, Z.; Diwan, A.D. Nociceptive pain assessed by the PainDETECT questionnaire may predict response to opioid treatment for chronic low back pain. Heliyon 2024, 10, e25834. [Google Scholar] [CrossRef]
  6. Kaplan, C.M.; Kelleher, E.; Irani, A.; Schrepf, A.; Clauw, D.J.; Harte, S.E. Deciphering nociplastic pain: Clinical features, risk factors and potential mechanisms. Nat. Rev. Neurol. 2024, 20, 347–363. [Google Scholar] [CrossRef]
  7. Rugnath, R.; Orzechowicz, C.; Newell, C.; Carullo, V.; Rugnath, A. A Literature Review: The Mechanisms and Treatment of Neuropathic Pain—A Brief Discussion. Biomedicines 2024, 12, 204. [Google Scholar] [CrossRef]
  8. van Hecke, O.; Austin, S.K.; Khan, R.A.; Smith, B.H.; Torrance, N. Neuropathic pain in the general population: A systematic review of epidemiological studies. Pain 2014, 155, 654–662. [Google Scholar] [CrossRef]
  9. Martins, J.P.; Marson, F.A.L. A narrative review of the complex panorama regarding chronic neuropathic pain mainly for the psychological issues. Heliyon 2024, 10, e38282. [Google Scholar] [CrossRef]
  10. Madariaga Muñoz, M.C.; Villegas Estévez, F.; Jiménez López, A.J.; Cabezón Álvarez, A.; Soler López, B. Evaluation of Quality of Life and Satisfaction of Patients with Neuropathic Pain and Breakthrough Pain: Economic Impact Based on Quality of Life. Pain Res. Treat. 2018, 2018, 5394021. [Google Scholar] [CrossRef]
  11. Cao, B.; Xu, Q.; Shi, Y.; Zhao, R.; Li, H.; Zheng, J.; Liu, F.; Wan, Y.; Wei, B. Pathology of pain and its implications for therapeutic interventions. Signal Transduct. Target. Ther. 2024, 9, 155. [Google Scholar] [CrossRef]
  12. Ghazisaeidi, S.; Muley, M.M.; Salter, M.W. Neuropathic Pain: Mechanisms, Sex Differences, and Potential Therapies for a Global Problem. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 565–583. [Google Scholar] [CrossRef]
  13. Bennett, G.J. What is spontaneous pain and who has it? J. Pain 2012, 13, 921–929. [Google Scholar] [CrossRef] [PubMed]
  14. Balzani, E.; Fanelli, A.; Malafoglia, V.; Tenti, M.; Ilari, S.; Corraro, A.; Muscoli, C.; Raffaeli, W. A Review of the Clinical and Therapeutic Implications of Neuropathic Pain. Biomedicines 2021, 9, 1239. [Google Scholar] [CrossRef]
  15. Bowsher, D. Dynamic mechanical allodynia in neuropathic pain. Pain 2005, 116, 164–165. [Google Scholar] [CrossRef]
  16. Jensen, T.S.; Finnerup, N.B. Allodynia and hyperalgesia in neuropathic pain: Clinical manifestations and mechanisms. Lancet Neurol. 2014, 13, 924–935. [Google Scholar] [CrossRef]
  17. Hamdan, A.; Galvez, R.; Katati, M. Shedding light on neuropathic pain: Current and emerging tools for diagnosis, screening, and quantification. SAGE Open Med. 2024, 12, 20503121231218985. [Google Scholar] [CrossRef]
  18. van Velzen, M.; Dahan, A.; Niesters, M. Neuropathic Pain: Challenges and Opportunities. Front. Pain Res. 2020, 1, 1. [Google Scholar] [CrossRef]
  19. Duan, Y.-W.; Chen, S.-X.; Li, Q.-Y.; Zang, Y. Neuroimmune Mechanisms Underlying Neuropathic Pain: The Potential Role of TNF-α-Necroptosis Pathway. Int. J. Mol. Sci. 2022, 23, 7191. [Google Scholar] [CrossRef] [PubMed]
  20. Clark, A.K.; Old, E.A.; Malcangio, M. Neuropathic pain and cytokines: Current perspectives. J. Pain Res. 2013, 6, 803–814. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, F.; Zhao, C.; Jing, Z.; Wang, Q.; Li, M.; Lu, B.; Huo, A.; Liang, W.; Hu, W.; Fu, X. The dual roles of chemokines in peripheral nerve injury and repair. Inflamm. Regen. 2025, 45, 11. [Google Scholar] [CrossRef]
  22. Kato, J.; Svensson, C.I. Role of extracellular damage-associated molecular pattern molecules (DAMPs) as mediators of persistent pain. Prog. Mol. Biol. Transl. Sci. 2015, 131, 251–279. [Google Scholar]
  23. Araldi, D.; Khomula, E.V.; Bonet, I.J.M.; Bogen, O.; Green, P.G.; Levine, J.D. Role of pattern recognition receptors in chemotherapy-induced neuropathic pain. Brain 2024, 147, 1025–1042. [Google Scholar] [CrossRef] [PubMed]
  24. Prado, J.; Westerink, R.H.S.; Popov-Celeketic, J.; Steen-Louws, C.; Pandit, A.; Versteeg, S.; van de Worp, W.; Kanters, D.H.A.J.; Reedquist, K.A.; Koenderman, L.; et al. Cytokine receptor clustering in sensory neurons with an engineered cytokine fusion protein triggers unique pain resolution pathways. Proc. Natl. Acad. Sci. USA 2021, 118, e2009647118. [Google Scholar] [CrossRef] [PubMed]
  25. White, F.A.; Feldman, P.; Miller, R.J. Chemokine signaling and the management of neuropathic pain. Mol. Interv. 2009, 9, 188–195. [Google Scholar] [CrossRef]
  26. Donnelly, C.R.; Andriessen, A.S.; Chen, G.; Wang, K.; Jiang, C.; Maixner, W.; Ji, R.R. Central Nervous System Targets: Glial Cell Mechanisms in Chronic Pain. Neurotherapeutics 2020, 17, 846–860. [Google Scholar] [CrossRef] [PubMed]
  27. García-Domínguez, M. NGF in Neuropathic Pain: Understanding Its Role and Therapeutic Opportunities. Curr. Issues Mol. Biol. 2025, 47, 93. [Google Scholar] [CrossRef]
  28. Chaudhari, A.; Padmar, J.; Awathale, S.; Goyal, S.; Nakhate, K.; Sherikar, A. From glial cells to pain pathways: ICAM-1 as a central player in neuroinflammation and neuropathy. Discov. Neurosci. 2025, 20, 5. [Google Scholar] [CrossRef]
  29. Bennett, D.L.; Clark, A.J.; Huang, J.; Waxman, S.G.; Dib-Hajj, S.D. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol. Rev. 2019, 99, 1079–1151. [Google Scholar] [CrossRef]
  30. Alles, S.R.A.; Smith, P.A. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. Front. Pain Res. 2021, 2, 750583. [Google Scholar] [CrossRef]
  31. Busserolles, J.; Tsantoulas, C.; Eschalier, A.; López García, J.A. Potassium channels in neuropathic pain: Advances, challenges, and emerging ideas. Pain 2016, 157 (Suppl. S1), S7–S14. [Google Scholar] [CrossRef]
  32. Yin, Y.; Yi, M.H.; Kim, D.W. Impaired Autophagy of GABAergic Interneurons in Neuropathic Pain. Pain Res. Manag. 2018, 2018, 9185368. [Google Scholar] [CrossRef]
  33. Imlach, W.L.; Bhola, R.F.; Mohammadi, S.A.; Christie, M.J. Glycinergic dysfunction in a subpopulation of dorsal horn interneurons in a rat model of neuropathic pain. Sci. Rep. 2016, 6, 37104. [Google Scholar] [CrossRef]
  34. Li, X.Y.; Wan, Y.; Tang, S.J.; Guan, Y.; Wei, F.; Ma, D. Maladaptive Plasticity and Neuropathic Pain. Neural Plast. 2016, 2016, 4842159. [Google Scholar] [CrossRef] [PubMed]
  35. Pawlik, K.; Mika, J. Targeting Members of the Chemokine Family as a Novel Approach to Treating Neuropathic Pain. Molecules 2023, 28, 5766. [Google Scholar] [CrossRef]
  36. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef] [PubMed]
  37. Jiang, B.C.; Liu, T.; Gao, Y.J. Chemokines in chronic pain: Cellular and molecular mechanisms and therapeutic potential. Pharmacol. Ther. 2020, 212, 107581. [Google Scholar] [CrossRef] [PubMed]
  38. Bajetto, A.; Bonavia, R.; Barbero, S.; Florio, T.; Schettini, G. Chemokines and their receptors in the central nervous system. Front. Neuroendocrinol. 2001, 22, 147–184. [Google Scholar] [CrossRef]
  39. Lin, Y.; Liu, S.; Sun, Y.; Chen, C.; Yang, S.; Pei, G.; Lin, M.; Yu, J.; Liu, X.; Wang, H.; et al. CCR5 and inflammatory storm. Ageing Res. Rev. 2024, 96, 102286. [Google Scholar] [CrossRef]
  40. Shen, Y.; Zhou, M.; Cai, D.; Filho, D.A.; Fernandes, G.; Cai, Y.; de Sousa, A.F.; Tian, M.; Kim, N.; Lee, J.; et al. CCR5 closes the temporal window for memory linking. Nature 2022, 606, 146–152. [Google Scholar] [CrossRef]
  41. Steffens, C.M.; Hope, T.J. Localization of CD4 and CCR5 in living cells. J. Virol. 2003, 77, 4985–4991. [Google Scholar] [CrossRef]
  42. Festa, B.P.; Siddiqi, F.H.; Jimenez-Sanchez, M.; Won, H.; Rob, M.; Djajadikerta, A.; Stamatakou, E.; Rubinsztein, D.C. Microglial-to-neuronal CCR5 signaling regulates autophagy in neurodegeneration. Neuron 2023, 111, 2021–2037.e12. [Google Scholar] [CrossRef]
  43. Dorf, M.E.; Berman, M.A.; Tanabe, S.; Heesen, M.; Luo, Y. Astrocytes express functional chemokine receptors. J. Neuroimmunol. 2000, 111, 109–121. [Google Scholar] [CrossRef]
  44. Zhang, P.; Wang, Y.; Xu, Y.; Li, X.; Yu, X.; Li, Y.; Zhu, D.; Luo, F.; Li, W.; Jin, X. Nasal-to-brain delivery of CCR5 antagonist for reshaping the dysregulated microglia-neuron axis and enhancing post-traumatic cognitive function. Biomaterials 2026, 324, 123479. [Google Scholar] [CrossRef] [PubMed]
  45. Oppermann, M. Chemokine receptor CCR5: Insights into structure, function, and regulation. Cell. Signal. 2004, 16, 1201–1210. [Google Scholar] [CrossRef]
  46. Zare-Bidaki, M.; Karimi-Googheri, M.; Hassanshahi, G.; Zainodini, N.; Arababadi, M.K. The frequency of CCR5 promoter polymorphisms and CCR5 Δ 32 mutation in Iranian populations. Iran. J. Basic Med. Sci. 2015, 18, 312–316. [Google Scholar] [PubMed]
  47. Saika, F.; Sato, T.; Nakabayashi, T.; Fukazawa, Y.; Hino, S.; Suzuki, K.; Kiguchi, N. Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury. Cells 2025, 14, 484. [Google Scholar] [CrossRef] [PubMed]
  48. Isaikina, P.; Tsai, C.J.; Dietz, N.; Pamula, F.; Grahl, A.; Goldie, K.N.; Guixà-González, R.; Branco, C.; Paolini-Bertrand, M.; Calo, N.; et al. Structural basis of the activation of the CC chemokine receptor 5 by a chemokine agonist. Sci. Adv. 2021, 7, eabg8685. [Google Scholar] [CrossRef]
  49. Jasinska, A.J.; 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. [Google Scholar] [CrossRef]
  50. Jensen, T.S. Anticonvulsants in neuropathic pain: Rationale and clinical evidence. Eur. J. Pain 2002, 6, 61–68. [Google Scholar] [CrossRef]
  51. Catalisano, G.; Campione, G.M.; Spurio, G.; Galvano, A.N.; di Villalba, C.P.; Giarratano, A.; Alongi, A.; Ippolito, M.; Cortegiani, A. Neuropathic pain, antidepressant drugs, and inflammation: A narrative review. J. Anesth. Analg. Crit. Care 2024, 4, 67. [Google Scholar] [CrossRef]
  52. McNicol, E.D.; Midbari, A.; Eisenberg, E. Opioids for neuropathic pain. Cochrane Database Syst. Rev. 2013, 2013, CD006146. [Google Scholar] [CrossRef]
  53. Cavalli, E.; Mammana, S.; Nicoletti, F.; Bramanti, P.; Mazzon, E. The neuropathic pain: An overview of the current treatment and future therapeutic approaches. Int. J. Immunopathol. Pharmacol. 2019, 33, 2058738419838383. [Google Scholar] [CrossRef] [PubMed]
  54. Piotrowska, A.; Kwiatkowski, K.; Rojewska, E.; Makuch, W.; Mika, J. Maraviroc reduces neuropathic pain through polarization of microglia and astroglia—Evidence from in vivo and in vitro studies. Neuropharmacology 2016, 108, 207–219. [Google Scholar] [CrossRef] [PubMed]
  55. Bober, A.; Piotrowska, A.; Pawlik, K.; Ciapała, K.; Maciuszek, M.; Makuch, W.; Mika, J. A New Application for Cenicriviroc, a Dual CCR2/CCR5 Antagonist, in the Treatment of Painful Diabetic Neuropathy in a Mouse Model. Int. J. Mol. Sci. 2024, 25, 7410. [Google Scholar] [CrossRef]
  56. Lee, Y.K.; Choi, D.Y.; Jung, Y.Y.; Yun, Y.W.; Lee, B.J.; Han, S.B.; Hong, J.T. Decreased pain responses of C-C chemokine receptor 5 knockout mice to chemical or inflammatory stimuli. Neuropharmacology 2013, 67, 57–65. [Google Scholar] [CrossRef]
  57. Zhou, P.; Zhang, R.; Xian, L.; Ning, L.; Lu, P.; Liu, Q.; Liu, M. Selection of sciatic nerve injury models: Implications for pathogenesis and treatment. Front. Neurol. 2025, 16, 1521941. [Google Scholar] [CrossRef] [PubMed]
  58. Lee, K.T.; Bulls, H.W.; Hoogland, A.I.; James, B.W.; Colon-Echevarria, C.B.; Jim, H.S.L. Chemotherapy-Induced Peripheral Neuropathy (CIPN): A Narrative Review and Proposed Theoretical Model. Cancers 2024, 16, 2571. [Google Scholar] [CrossRef]
  59. Sun, C.F.; Lin, Y.H.; Ling, G.X.; Gao, H.J.; Feng, X.Z.; Sun, C.Q. Systematic review and critical appraisal of predictive models for diabetic peripheral neuropathy: Existing challenges and proposed enhancements. World J. Diabetes 2025, 16, 101310. [Google Scholar] [CrossRef]
  60. Faivre, N.; Verollet, C.; Dumas, F. The chemokine receptor CCR5: Multi-faceted hook for HIV-1. Retrovirology 2024, 21, 2. [Google Scholar] [CrossRef]
  61. Parmentier, M. CCR5 and HIV Infection, a View from Brussels. Front. Immunol. 2015, 6, 295. [Google Scholar] [CrossRef]
  62. Zhang, H.; Chen, K.; Tan, Q.; Shao, Q.; Han, S.; Zhang, C.; Yi, C.; Chu, X.; Zhu, Y.; Xu, Y.; et al. Structural basis for chemokine recognition and receptor activation of chemokine receptor CCR5. Nat. Commun. 2021, 12, 4151. [Google Scholar] [CrossRef]
  63. Chain, B.; Arnold, J.; Akthar, S.; Brandt, M.; Davis, D.; Noursadeghi, M.; Lapp, T.; Ji, C.; Sankuratri, S.; Zhang, Y.; et al. A Linear Epitope in the N-Terminal Domain of CCR5 and Its Interaction with Antibody. PLoS ONE 2015, 10, e0128381. [Google Scholar] [CrossRef][Green Version]
  64. Itell, H.L.; Guenthoer, J.; Humes, D.; Baumgarten, N.E.; Overbaugh, J. Host cell glycosylation selects for infection with CCR5- versus CXCR4-tropic HIV-1. Nat. Microbiol. 2024, 9, 2985–2996. [Google Scholar] [CrossRef] [PubMed]
  65. Bannert, N.; Craig, S.; Farzan, M.; Sogah, D.; Santo, N.V.; Choe, H.; Sodroski, J. 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, 1661–1674. [Google Scholar] [CrossRef]
  66. Samson, M.; LaRosa, G.; Libert, F.; Paindavoine, P.; Detheux, M.; Vassart, G.; Parmentier, M. The second extracellular loop of CCR5 is the major determinant of ligand specificity. J. Biol. Chem. 1997, 272, 24934–24941. [Google Scholar] [CrossRef] [PubMed]
  67. Steen, A.; Thiele, S.; Guo, D.; Hansen, L.S.; Frimurer, T.M.; Rosenkilde, M.M. Biased and constitutive signaling in the CC-chemokine receptor CCR5 by manipulating the interface between transmembrane helices 6 and 7. J. Biol. Chem. 2013, 288, 12511–12521. [Google Scholar] [CrossRef]
  68. Lin, Y.L.; Mettling, C.; Portalès, P.; Réant, B.; Robert-Hebmann, V.; Reynes, J.; Clot, J.; Corbeau, P. The efficiency of R5 HIV-1 infection is determined by CD4 T-cell surface CCR5 density through Gαi-protein signalling. AIDS 2006, 20, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  69. Zhao, J.; Ma, L.; Wu, Y.L.; Wang, P.; Hu, W.; Pei, G. Chemokine receptor CCR5 functionally couples to inhibitory G proteins and undergoes desensitization. J. Cell. Biochem. 1998, 71, 36–45. [Google Scholar] [CrossRef]
  70. Spadaro, F.; Cecchetti, S.; Purificato, C.; Sabbatucci, M.; Podo, F.; Ramoni, C.; Gessani, S.; Fantuzzi, L. Nuclear phosphoinositide-specific phospholipase C β1 controls cytoplasmic CCL2 mRNA levels in HIV-1 gp120-stimulated primary human macrophages. PLoS ONE 2013, 8, e59705. [Google Scholar] [CrossRef]
  71. Hang, L.H.; Li, S.N.; Dan, X.; Shu, W.W.; Luo, H.; Shao, D.H. Involvement of Spinal CCR5/PKCγ Signaling Pathway in the Maintenance of Cancer-Induced Bone Pain. Neurochem. Res. 2017, 42, 563–571. [Google Scholar] [CrossRef]
  72. Popik, W.; Pitha, P.M. Early activation of mitogen-activated protein kinase kinase, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and c-Jun N-terminal kinase in response to binding of simian immunodeficiency virus to Jurkat T cells expressing CCR5 receptor. Virology 1998, 252, 210–217. [Google Scholar]
  73. Zhu, B.; Wei, R.; Li, X.; Bi, Q. Targeting CCL5 Attenuates Fibrosis via Activation of PI3k/Akt Signaling Axis After Glaucoma Filtration Surgery. Curr. Eye Res. 2025, 50, 394–404. [Google Scholar] [CrossRef]
  74. Lucera, M.B.; Fleissner, Z.; Tabler, C.O.; Schlatzer, D.M.; Troyer, Z.; Tilton, J.C. 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. [Google Scholar] [CrossRef]
  75. Venkatesan, S.; Petrovic, A.; Locati, M.; Kim, Y.O.; Weissman, D.; Murphy, P.M. A membrane-proximal basic domain and cysteine cluster in the C-terminal tail of CCR5 constitute a bipartite motif critical for cell surface expression. J. Biol. Chem. 2001, 276, 40133–40145. [Google Scholar] [CrossRef]
  76. Stambouli, N.; Dridi, M.; Wei, N.N.; Jlizi, A.; Bouraoui, A.; Elgaaied, A.B. Structural insight into the binding complex: β-arrestin/CCR5 complex. J. Biomol. Struct. Dyn. 2014, 32, 866–875. [Google Scholar] [CrossRef] [PubMed]
  77. Petrovic, I.; Desai, S.; Isaikina, P.; Abiko, L.A.; Spang, A.; Grzesiek, S. A high-resolution analysis of arrestin2 interactions responsible for CCR5 endocytosis. eLife 2025, 14, RP106839. [Google Scholar]
  78. Mueller, A.; Kelly, E.; Strange, P.G. Pathways for internalization and recycling of the chemokine receptor CCR5. Blood 2002, 99, 785–791. [Google Scholar] [CrossRef]
  79. Bauss, J.; Morris, M.; Shankar, R.; Olivero, R.; Buck, L.N.; Stenger, C.L.; Hinds, D.; Mills, J.; Eby, A.; Zagorski, J.W.; et al. CCR5 and Biological Complexity: The Need for Data Integration and Educational Materials to Address Genetic/Biological Reductionism at the Interface of Ethical, Legal, and Social Implications. Front. Immunol. 2021, 12, 790041. [Google Scholar] [CrossRef] [PubMed]
  80. Verma, M.K.; Shakya, S. Genetic variation in the chemokine receptor 5 gene and course of HIV infection; review on genetics and immunological aspect. Genes Dis. 2020, 8, 475–483. [Google Scholar] [CrossRef] [PubMed]
  81. Pekkoc-Uyanik, K.C.; Todurga-Seven, Z.G.; Shahzadi, A.; Sonmez, H.; Mercan, S.; Mete, B.; Tabak, F. Next-generation sequencing of CCR5, CXCR4, and IFNAR1 variants in relation to HIV-1 disease progression and ART response. Sci. Rep. 2025, 15, 26511. [Google Scholar] [CrossRef]
  82. Obrien, S.J. Legacy of a magic gene-CCR5-∆32: From discovery to clinical benefit in a generation. Proc. Natl. Acad. Sci. USA 2024, 121, e2321907121. [Google Scholar] [CrossRef]
  83. Ravn, K.; Cobuccio, L.; Muktupavela, R.A.; Meisner, J.; Danielsen, L.S.; Benros, M.E.; Korneliussen, T.S.; Sikora, M.; Willerslev, E.; Allentoft, M.E.; et al. Tracing the evolutionary history of the CCR5delta32 deletion via ancient and modern genomes. Cell 2025, 188, 3679–3695.e16. [Google Scholar] [CrossRef]
  84. Balotta, C.; Bagnarelli, P.; Violin, M.; Ridolfo, A.L.; Zhou, D.; Berlusconi, A.; Corvasce, S.; Corbellino, M.; Clementi, M.; Clerici, M.; et al. Homozygous delta 32 deletion of the CCR-5 chemokine receptor gene in an HIV-1-infected patient. AIDS 1997, 11, F67–F71. [Google Scholar] [CrossRef]
  85. Liu, S.; Kong, C.; Wu, J.; Ying, H.; Zhu, H. Effect of CCR5-Δ32 heterozygosity on HIV-1 susceptibility: A meta-analysis. PLoS ONE 2012, 7, e35020. [Google Scholar] [CrossRef] [PubMed]
  86. Glass, W.G.; McDermott, D.H.; Lim, J.K.; Lekhong, S.; Yu, S.F.; Frank, W.A.; Pape, J.; Cheshier, R.C.; Murphy, P.M. CCR5 deficiency increases risk of symptomatic West Nile virus infection. J. Exp. Med. 2006, 203, 35–40. [Google Scholar] [CrossRef] [PubMed]
  87. Dore, M.P.; Errigo, A.; Merola, E.; Pes, G.M. Role of ACE1, ACE2, and CCR5-Δ32 Polymorphisms in the Transmission of SARS-CoV-2 to Intimate Contacts. Biology 2025, 14, 587. [Google Scholar] [CrossRef]
  88. Goel, V.; Bose, P.D.; Sarma, M.P.; Hazam, R.K.; Das, B.C.; Gondal, R.; Kar, P. Chemokine receptor 5 (CCR5) polymorphism in chronic hepatitis B patients treated with three different nucleos(t)ide analogues. Indian J. Med. Res. 2013, 137, 1208–1209. [Google Scholar]
  89. Zapico, I.; Coto, E.; Rodríguez, A.; Alvarez, C.; Torre, J.C.; Alvarez, V. CCR5 (chemokine receptor-5) DNA-polymorphism influences the severity of rheumatoid arthritis. Genes Immun. 2000, 1, 288–289. [Google Scholar] [CrossRef]
  90. Rzeszotarska, E.; Sowinska, A.; Stypinska, B.; Walczuk, E.; Wajda, A.; Lutkowska, A.; Felis-Giemza, A.; Olesinska, M.; Puszczewicz, M.; Majewski, D.; et al. The Role of MECP2 and CCR5 Polymorphisms on the Development and Course of Systemic Lupus Erythematosus. Biomolecules 2020, 10, 494. [Google Scholar] [CrossRef] [PubMed]
  91. Glass, W.G.; Lim, J.K.; Cholera, R.; Pletnev, A.G.; Gao, J.L.; Murphy, P.M. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection. J. Exp. Med. 2005, 202, 1087–1098. [Google Scholar] [CrossRef] [PubMed]
  92. Jin, C.; Zhang, F.; Luo, H.; Li, B.; Jiang, X.; Pirozzi, C.J.; Liang, C.; Zhang, M. The CCL5/CCR5/SHP2 axis sustains Stat1 phosphorylation and activates NF-κB signaling promoting M1 macrophage polarization and exacerbating chronic prostatic inflammation. Cell Commun. Signal. 2024, 22, 584. [Google Scholar] [CrossRef] [PubMed]
  93. Alonzo, F.; Torres, V.J. Staphylococcus aureus and CCR5: Unveiling commonalities in host-pathogen interactions and potential treatment strategies. Future Microbiol. 2013, 8, 425–428. [Google Scholar] [CrossRef] [PubMed]
  94. Westmoreland, S.V.; Alvarez, X.; deBakker, C.; Aye, P.; Wilson, M.L.; Williams, K.C.; Lackner, A.A. Developmental expression patterns of CCR5 and CXCR4 in the rhesus macaque brain. J. Neuroimmunol. 2002, 122, 146–158. [Google Scholar] [CrossRef]
  95. Lynch, E.A.; Heijens, C.A.; Horst, N.F.; Center, D.M.; Cruikshank, W.W. Cutting edge: IL-16/CD4 preferentially induces Th1 cell migration: Requirement of CCR5. J. Immunol. 2003, 171, 4965–4968. [Google Scholar] [CrossRef]
  96. Urszula, Ł.; Ulana, J.; Bartosz, S.; Maja, O.; Małgorzata, M.; Monika, R.S. Exploring CCR5+ T regulatory cell subset dysfunction in type 1 diabetes patients: Implications for immune regulation. Immunol. Res. 2024, 72, 1061–1070. [Google Scholar] [CrossRef]
  97. Desmetz, C.; Lin, Y.L.; Mettling, C.; Portalès, P.; Rabesandratana, H.; Clot, J.; Corbeau, P. The strength of the chemotactic response to a CCR5 binding chemokine is determined by the level of cell surface CCR5 density. Immunology 2006, 119, 551–561. [Google Scholar] [CrossRef]
  98. Seo, I.H.; Eun, H.S.; Kim, J.K.; Lee, H.; Jeong, S.; Choi, S.J.; Lee, J.; Lee, B.S.; Kim, S.H.; Rou, W.S.; et al. IL-15 enhances CCR5-mediated migration of memory CD8+ T cells by upregulating CCR5 expression in the absence of TCR stimulation. Cell Rep. 2021, 36, 109438. [Google Scholar] [CrossRef]
  99. Ferrero, M.R.; Tavares, L.P.; Garcia, C.C. The Dual Role of CCR5 in the Course of Influenza Infection: Exploring Treatment Opportunities. Front. Immunol. 2022, 12, 826621. [Google Scholar] [CrossRef]
  100. Renovato-Martins, M.; Matheus, M.E.; de Andrade, I.R.; Moraes, J.A.; da Silva, S.V.; Citelli Dos Reis, M.; de Souza, A.A.; da Silva, C.C.; Bouskela, E.; Barja-Fidalgo, C. Microparticles derived from obese adipose tissue elicit a pro-inflammatory phenotype of CD16+, CCR5+ and TLR8+ monocytes. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 139–151. [Google Scholar] [CrossRef]
  101. Shen, R.; Meng, G.; Ochsenbauer, C.; Clapham, P.R.; Grams, J.; Novak, L.; Kappes, J.C.; Smythies, L.E.; Smith, P.D. Stromal down-regulation of macrophage CD4/CCR5 expression and NF-κB activation mediates HIV-1 non-permissiveness in intestinal macrophages. PLoS Pathog. 2011, 7, e1002060. [Google Scholar] [CrossRef]
  102. Bartneck, M.; Koppe, C.; Fech, V.; Warzecha, K.T.; Kohlhepp, M.; Huss, S.; Weiskirchen, R.; Trautwein, C.; Luedde, T.; Tacke, F. Roles of CCR2 and CCR5 for Hepatic Macrophage Polarization in Mice with Liver Parenchymal Cell-Specific NEMO Deletion. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 327–347. [Google Scholar] [CrossRef]
  103. Matsuda, M.; Shimora, H.; Nagatani, Y.; Nishikawa, K.; Takamori, I.; Haguchi, T.; Kitatani, K.; Kaminuma, O.; Nabe, T. Involvement of CCR5 on interstitial macrophages in the development of lung fibrosis in severe asthma. Int. Immunopharmacol. 2024, 135, 112331. [Google Scholar] [CrossRef] [PubMed]
  104. Feng, M.; Zhou, S.; Yu, Y.; Su, Q.; Li, X.; Lin, W. Regulation of the Migration of Distinct Dendritic Cell Subsets. Front. Cell Dev. Biol. 2021, 9, 635221. [Google Scholar] [CrossRef]
  105. Santos, J.; Wang, P.; Shemesh, A.; Liu, F.; Tsao, T.; Aguilar, O.A.; Cleary, S.J.; Singer, J.P.; Gao, Y.; Hays, S.R.; et al. CCR5 drives NK cell-associated airway damage in pulmonary ischemia-reperfusion injury. JCI Insight 2023, 8, e173716. [Google Scholar] [CrossRef]
  106. Zhang, Z.; Wang, Q.; Yao, J.; Zhou, X.; Zhao, J.; Zhang, X.; Dong, J.; Liao, L. Chemokine Receptor 5, a Double-Edged Sword in Metabolic Syndrome and Cardiovascular Disease. Front. Pharmacol. 2020, 11, 146. [Google Scholar] [CrossRef]
  107. Costa, R.M.; Cerqueira, D.M.; Bruder-Nascimento, A.; Alves, J.V.; Awata, W.M.C.; Singh, S.; Kufner, A.; Prado, D.S.; Johny, E.; Cifuentes-Pagano, E.; et al. Role of the CCL5 and Its Receptor, CCR5, in the Genesis of Aldosterone-Induced Hypertension, Vascular Dysfunction, and End-Organ Damage. Hypertension 2024, 81, 776–786. [Google Scholar] [CrossRef] [PubMed]
  108. Ohta-Ogo, K.; Ishibashi-Ueda, H.; Ogo, T.; Ikeda, Y.; Inagaki, T.; Matsumoto, M.; Amemiya, K.; Nakaoka, Y.; Hatakeyama, K. Recruitment of CCR5-positive T cells in pulmonary vascular lesions in idiopathic- and connective tissue disease associated- pulmonary arterial hypertension. Eur. Heart J. 2024, 45, ehae666.2165. [Google Scholar] [CrossRef]
  109. Cowell, R.M.; Xu, H.; Parent, J.M.; Silverstein, F.S. Microglial expression of chemokine receptor CCR5 during rat forebrain development and after perinatal hypoxia-ischemia. J. Neuroimmunol. 2006, 173, 155–165. [Google Scholar] [CrossRef] [PubMed]
  110. Trebst, C.; Staugaitis, S.M.; Tucky, B.; Wei, T.; Suzuki, K.; Aldape, K.D.; Pardo, C.A.; Troncoso, J.; Lassmann, H.; Ransohoff, R.M. Chemokine receptors on infiltrating leucocytes in inflammatory pathologies of the central nervous system (CNS). Neuropathol. Appl. Neurobiol. 2003, 29, 584–595. [Google Scholar] [CrossRef]
  111. Hamid, R.; Alaziz, M.; Mahal, A.S.; Ashton, A.W.; Halama, N.; Jaeger, D.; Jiao, X.; Pestell, R.G. The Role and Therapeutic Targeting of CCR5 in Breast Cancer. Cells 2023, 12, 2237. [Google Scholar] [CrossRef] [PubMed]
  112. Xiong, Z.; Yu, S.L.; Xie, Z.X.; Zhuang, R.L.; Peng, S.R.; Wang, Q.; Gao, Z.; Li, B.H.; Xie, J.J.; Huang, H.; et al. Cancer-associated fibroblasts promote enzalutamide resistance and PD-L1 expression in prostate cancer through CCL5-CCR5 paracrine axis. iScience 2024, 27, 109674. [Google Scholar] [CrossRef]
  113. Battaglin, F.; Baca, Y.; Millstein, J.; Yang, Y.; Xiu, J.; Arai, H.; Wang, J.; Ou, F.S.; Innocenti, F.; Mumenthaler, S.M.; et al. CCR5 and CCL5 gene expression in colorectal cancer: Comprehensive profiling and clinical value. J. Immunother. Cancer 2024, 12, e007939. [Google Scholar] [CrossRef]
  114. Huang, H.; Zepp, M.; Georges, R.B.; Jarahian, M.; Kazemi, M.; Eyol, E.; Berger, M.R. The CCR5 antagonist maraviroc causes remission of pancreatic cancer liver metastasis in nude rats based on cell cycle inhibition and apoptosis induction. Cancer Lett. 2020, 474, 82–93. [Google Scholar] [CrossRef]
  115. Mencarelli, A.; Graziosi, L.; Renga, B.; Cipriani, S.; D’Amore, C.; Francisci, D.; Bruno, A.; Baldelli, F.; Donini, A.; Fiorucci, S. CCR5 Antagonism by Maraviroc Reduces the Potential for Gastric Cancer Cell Dissemination. Transl. Oncol. 2013, 6, 784–793. [Google Scholar] [CrossRef]
  116. Korbecki, J.; Bosiacki, M.; Stasiak, P.; Snarski, E.; Brodowska, A.; Chlubek, D.; Baranowska-Bosiacka, I. Clinical Aspects and Significance of β-Chemokines, γ-Chemokines, and δ-Chemokines in Molecular Cancer Processes in Acute Myeloid Leukemia (AML) and Myelodysplastic Neoplasms (MDS). Cancers 2024, 16, 3246. [Google Scholar] [CrossRef]
  117. Menu, E.; De Leenheer, E.; De Raeve, H.; Coulton, L.; Imanishi, T.; Miyashita, K.; Van Valckenborgh, E.; Van Riet, I.; Van Camp, B.; Horuk, R.; et al. Role of CCR1 and CCR5 in homing and growth of multiple myeloma and in the development of osteolytic lesions: A study in the 5TMM model. Clin. Exp. Metastasis 2006, 23, 291–300. [Google Scholar] [CrossRef]
  118. Aldinucci, D.; Borghese, C.; Casagrande, N. The CCL5/CCR5 Axis in Cancer Progression. Cancers 2020, 12, 1765. [Google Scholar] [CrossRef]
  119. Lopalco, L. CCR5: From Natural Resistance to a New Anti-HIV Strategy. Viruses 2010, 2, 574–600. [Google Scholar] [CrossRef] [PubMed]
  120. Bender, E.C.; Tareq, H.S.; Suggs, L.J. Inflammation: A matter of immune cell life and death. Npj Biomed. Innov. 2025, 2, 7. [Google Scholar] [CrossRef]
  121. Loetscher, P.; Uguccioni, M.; Bordoli, L.; Baggiolini, M.; Moser, B.; Chizzolini, C.; Dayer, J.M. CCR5 is characteristic of Th1 lymphocytes. Nature 1998, 391, 344–345. [Google Scholar] [CrossRef]
  122. Kroetz, D.N.; Deepe, G.S., Jr. CCR5 dictates the equilibrium of proinflammatory IL-17+ and regulatory Foxp3+ T cells in fungal infection. J. Immunol. 2010, 184, 5224–5231. [Google Scholar] [CrossRef]
  123. Rawat, K.; Tewari, A.; Li, X.; Mara, A.B.; King, W.T.; Gibbings, S.L.; Nnam, C.F.; Kolling, F.W.; Lambrecht, B.N.; Jakubzick, C.V. CCL5-producing migratory dendritic cells guide CCR5+ monocytes into the draining lymph nodes. J. Exp. Med. 2023, 220, e20222129. [Google Scholar] [CrossRef]
  124. Martín-Leal, A.; Blanco, R.; Casas, J.; Sáez, M.E.; Rodríguez-Bovolenta, E.; de Rojas, I.; Drechsler, C.; Real, L.M.; Fabrias, G.; Ruíz, A.; et al. CCR5 deficiency impairs CD4+ T-cell memory responses and antigenic sensitivity through increased ceramide synthesis. EMBO J. 2020, 39, e104749. [Google Scholar] [CrossRef]
  125. Foppiani, E.; Burnham, A.J.; Daley-Bauer, L.; Horwitz, E. Integrin α4 and CCR5 on IFNγ-activated MSC determine biodistribution to critical sites for aGVHD prophylaxis in mice. Cytotherapy 2025, 27, S11. [Google Scholar] [CrossRef]
  126. Blanco, R.; Gómez de Cedrón, M.; Gámez-Reche, L.; Martín-Leal, A.; González-Martín, A.; Lacalle, R.A.; Ramírez de Molina, A.; Mañes, S. The Chemokine Receptor CCR5 Links Memory CD4+ T Cell Metabolism to T Cell Antigen Receptor Nanoclustering. Front. Immunol. 2021, 12, 722320. [Google Scholar] [CrossRef] [PubMed]
  127. Schweneker, M.; Bachmann, A.S.; Moelling, K. The HIV-1 co-receptor CCR5 binds to alpha-catenin, a component of the cellular cytoskeleton. Biochem. Biophys. Res. Commun. 2004, 325, 751–757. [Google Scholar] [CrossRef] [PubMed]
  128. Sorce, S.; Myburgh, R.; Krause, K.H. The chemokine receptor CCR5 in the central nervous system. Prog. Neurobiol. 2011, 93, 297–311. [Google Scholar] [CrossRef]
  129. Liu, J.; Guo, Y.; Zhang, C.; Zeng, Y.; Luo, Y.; Wang, G. Clearance Systems in the Brain, From Structure to Function. Front. Cell. Neurosci. 2022, 15, 729706. [Google Scholar] [CrossRef]
  130. Louboutin, J.P.; Chekmasova, A.; Marusich, E.; Agrawal, L.; Strayer, D.S. Role of CCR5 and its ligands in the control of vascular inflammation and leukocyte recruitment required for acute excitotoxic seizure induction and neural damage. FASEB J. 2011, 25, 737–753. [Google Scholar] [CrossRef]
  131. Mohamed, H.; Gurrola, T.; Berman, R.; Collins, M.; Sariyer, I.K.; Nonnemacher, M.R.; Wigdahl, B. Targeting CCR5 as a Component of an HIV-1 Therapeutic Strategy. Front. Immunol. 2022, 12, 816515. [Google Scholar] [CrossRef] [PubMed]
  132. Raja, A.; Venturi, M.; Kwong, P.; Sodroski, J. CD4 binding site antibodies inhibit human immunodeficiency virus gp120 envelope glycoprotein interaction with CCR5. J. Virol. 2003, 77, 713–718. [Google Scholar] [CrossRef]
  133. Chen, B. Molecular Mechanism of HIV-1 Entry. Trends Microbiol. 2019, 27, 878–891. [Google Scholar] [CrossRef]
  134. Guan, Y. The first structure of HIV-1 gp120 with CD4 and CCR5 receptors. Cell Biosci. 2019, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  135. Bruxelle, J.-F.; Trattnig, N.; Mureithi, M.W.; Landais, E.; Pantophlet, R. HIV-1 Entry and Prospects for Protecting against Infection. Microorganisms 2021, 9, 228. [Google Scholar] [CrossRef]
  136. Bennetts, B.H.; Teutsch, S.M.; Buhler, M.M.; Heard, R.N.; Stewart, G.J. The CCR5 deletion mutation fails to protect against multiple sclerosis. Hum. Immunol. 1997, 58, 52–59. [Google Scholar] [CrossRef]
  137. Takeuchi, T.; Kameda, H. What is the future of CCR5 antagonists in rheumatoid arthritis? Arthritis Res. Ther. 2012, 14, 114. [Google Scholar] [CrossRef][Green Version]
  138. Schauren, J.D.S.; de Oliveira, A.H.; Consiglio, C.R.; Monticielo, O.A.; Xavier, R.M.; Nunes, N.S.; Ellwanger, J.H.; Chies, J.A.B. CCR5 promoter region polymorphisms in systemic lupus erythematosus. Int. J. Immunogenet. 2024, 51, 20–31. [Google Scholar] [CrossRef]
  139. Ishida, Y.; Kimura, A.; Kuninaka, Y.; Inui, M.; Matsushima, K.; Mukaida, N.; Kondo, T. Pivotal role of the CCL5/CCR5 interaction for recruitment of endothelial progenitor cells in mouse wound healing. J. Clin. Investig. 2012, 122, 711–721. [Google Scholar] [CrossRef] [PubMed]
  140. Pesaresi, M.; Bonilla-Pons, S.A.; Sebastian-Perez, R.; Di Vicino, U.; Alcoverro-Bertran, M.; Michael, R.; Cosma, M.P. The Chemokine Receptors Ccr5 and Cxcr6 Enhance Migration of Mesenchymal Stem Cells into the Degenerating Retina. Mol. Ther. 2021, 29, 804–821. [Google Scholar] [CrossRef]
  141. Zhuang, Y.; Zhao, X.; Yuan, B.; Zeng, Z.; Chen, Y. Blocking the CCL5-CCR5 Axis Using Maraviroc Promotes M1 Polarization of Macrophages Cocultured with Irradiated Hepatoma Cells. J. Hepatocell. Carcinoma 2021, 8, 599–611. [Google Scholar] [CrossRef] [PubMed]
  142. Kiseleva, V.; Vishnyakova, P.; Elchaninov, A.; Fatkhudinov, T.; Sukhikh, G. Biochemical and molecular inducers and modulators of M2 macrophage polarization in clinical perspective. Int. Immunopharmacol. 2023, 122, 110583. [Google Scholar] [CrossRef] [PubMed]
  143. Claiborne, D.T.; Detwiler, Z.; Docken, S.S.; Borland, T.D.; Cromer, D.; Simkhovich, A.; Ophinni, Y.; Okawa, K.; Bateson, T.; Chen, T.; et al. High frequency CCR5 editing in human hematopoietic stem progenitor cells protects xenograft mice from HIV infection. Nat. Commun. 2025, 16, 446. [Google Scholar] [CrossRef]
  144. McDermott, D.H.; Conway, S.E.; Wang, T.; Ricklefs, S.M.; Agovi, M.A.; Porcella, S.F.; Tran, H.T.; Milford, E.; Spellman, S.; Abdi, R. Donor and recipient chemokine receptor CCR5 genotype is associated with survival after bone marrow transplantation. Blood 2010, 115, 2311–2318. [Google Scholar] [CrossRef]
  145. Mende, N.; Laurenti, E. Hematopoietic stem and progenitor cells outside the bone marrow: Where, when, and why. Exp. Hematol. 2021, 104, 9–16. [Google Scholar] [CrossRef] [PubMed]
  146. Seyfried, A.N.; McCabe, A.; Smith, J.N.P.; Calvi, L.M.; MacNamara, K.C. CCR5 maintains macrophages in the bone marrow and drives hematopoietic failure in a mouse model of severe aplastic anemia. Leukemia 2021, 35, 3139–3151. [Google Scholar] [CrossRef]
  147. Robertson, P.; Means, T.K.; Luster, A.D.; Scadden, D.T. CXCR4 and CCR5 mediate homing of primitive bone marrow-derived hematopoietic cells to the postnatal thymus. Exp. Hematol. 2006, 34, 308–319. [Google Scholar] [CrossRef]
  148. Piryani, S.O.; Kam, A.Y.F.; Vu, U.T.; Chao, N.J.; Doan, P.L. CCR5 Signaling Promotes Murine and Human Hematopoietic Regeneration following Ionizing Radiation. Stem Cell Rep. 2019, 13, 76–90. [Google Scholar] [CrossRef]
  149. Sáez-Cirión, A.; Mamez, A.C.; Avettand-Fenoel, V.; Nabergoj, M.; Passaes, C.; Thoueille, P.; Decosterd, L.; Hentzien, M.; Perdomo-Celis, F.; Salgado, M.; et al. Sustained HIV remission after allogeneic hematopoietic stem cell transplantation with wild-type CCR5 donor cells. Nat. Med. 2024, 30, 3544–3554. [Google Scholar] [CrossRef]
  150. García-Domínguez, M. Injury-Driven Structural and Molecular Modifications in Nociceptors. Biology 2025, 14, 788. [Google Scholar] [CrossRef]
  151. Kocot-Kępska, M.; Zajączkowska, R.; Mika, J.; Wordliczek, J.; Dobrogowski, J.; Przeklasa-Muszyńska, A. Peripheral Mechanisms of Neuropathic Pain—The Role of Neuronal and Non-Neuronal Interactions and Their Implications for Topical Treatment of Neuropathic Pain. Pharmaceuticals 2021, 14, 77. [Google Scholar] [CrossRef]
  152. Malcangio, M. Role of the immune system in neuropathic pain. Scand. J. Pain 2019, 20, 33–37. [Google Scholar] [CrossRef] [PubMed]
  153. Bless, N.M.; Huber-Lang, M.; Guo, R.F.; Warner, R.L.; Schmal, H.; Czermak, B.J.; Shanley, T.P.; Crouch, L.D.; Lentsch, A.B.; Sarma, V.; et al. Role of CC chemokines (macrophage inflammatory protein-1 beta, monocyte chemoattractant protein-1, RANTES) in acute lung injury in rats. J. Immunol. 2000, 164, 2650–2659. [Google Scholar] [CrossRef]
  154. Ajuebor, M.N.; Hogaboam, C.M.; Kunkel, S.L.; Proudfoot, A.E.; Wallace, J.L. The chemokine RANTES is a crucial mediator of the progression from acute to chronic colitis in the rat. J. Immunol. 2001, 166, 552–558. [Google Scholar] [CrossRef]
  155. Kiguchi, N.; Maeda, T.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. Macrophage inflammatory protein-1alpha mediates the development of neuropathic pain following peripheral nerve injury through interleukin-1beta up-regulation. Pain 2010, 149, 305–315. [Google Scholar] [CrossRef]
  156. Becerra, L.; Bishop, J.; Barmettler, G.; Kainz, V.; Burstein, R.; Borsook, D. Brain network alterations in the inflammatory soup animal model of migraine. Brain Res. 2017, 1660, 36–46. [Google Scholar] [CrossRef] [PubMed]
  157. Köster, P.A.; Leipold, E.; Tigerholm, J.; Maxion, A.; Namer, B.; Stiehl, T.; Lampert, A. Nociceptor sodium channels shape subthreshold phase, upstroke, and shoulder of action potentials. J. Gen. Physiol. 2025, 157, e202313526. [Google Scholar] [CrossRef]
  158. Antunes, F.T.T.; Campos, M.M.; Carvalho, V.d.P.R.; da Silva Junior, C.A.; Magno, L.A.V.; de Souza, A.H.; Gomez, M.V. Current Drug Development Overview: Targeting Voltage-Gated Calcium Channels for the Treatment of Pain. Int. J. Mol. Sci. 2023, 24, 9223. [Google Scholar] [CrossRef]
  159. Mickle, A.D.; Shepherd, A.J.; Mohapatra, D.P. Nociceptive TRP Channels: Sensory Detectors and Transducers in Multiple Pain Pathologies. Pharmaceuticals 2016, 9, 72. [Google Scholar] [CrossRef]
  160. Rivera-Arconada, I.; Baccei, M.L.; López-García, J.A.; Bardoni, R. An electrophysiologist’s guide to dorsal horn excitability and pain. Front. Cell. Neurosci. 2025, 19, 1548252. [Google Scholar] [CrossRef] [PubMed]
  161. Sahbaie, P.; Shi, X.; Guo, T.Z.; Qiao, Y.; Yeomans, D.C.; Kingery, W.S.; Clark, D.J. Role of substance P signaling in enhanced nociceptive sensitization and local cytokine production after incision. Pain 2009, 145, 341–349. [Google Scholar] [CrossRef]
  162. Büttner, R.; Schulz, A.; Reuter, M.; Akula, A.K.; Mindos, T.; Carlstedt, A.; Riecken, L.B.; Baader, S.L.; Bauer, R.; Morrison, H. Inflammaging impairs peripheral nerve maintenance and regeneration. Aging Cell 2018, 17, e12833. [Google Scholar] [CrossRef]
  163. Zhang, F.; Vadakkan, K.I.; Kim, S.S.; Wu, L.J.; Shang, Y.; Zhuo, M. Selective activation of microglia in spinal cord but not higher cortical regions following nerve injury in adult mouse. Mol. Pain 2008, 4, 15. [Google Scholar] [CrossRef]
  164. Jung, H.; Bhangoo, S.; Banisadr, G.; Freitag, C.; Ren, D.; White, F.A.; Miller, R.J. Visualization of chemokine receptor activation in transgenic mice reveals peripheral activation of CCR2 receptors in states of neuropathic pain. J. Neurosci. 2009, 29, 8051–8062. [Google Scholar] [CrossRef] [PubMed]
  165. He, X.H.; Zang, Y.; Chen, X.; Pang, R.P.; Xu, J.T.; Zhou, X.; Wei, X.H.; Li, Y.Y.; Xin, W.J.; Qin, Z.H.; et al. TNF-α contributes to up-regulation of Nav1.3 and Nav1.8 in DRG neurons following motor fiber injury. Pain 2010, 151, 266–279. [Google Scholar] [CrossRef] [PubMed]
  166. Stemkowski, P.L.; Noh, M.C.; Chen, Y.; Smith, P.A. Increased excitability of medium-sized dorsal root ganglion neurons by prolonged interleukin-1β exposure is K+ channel dependent and reversible. J. Physiol. 2015, 593, 3739–3755. [Google Scholar] [CrossRef]
  167. García-Domínguez, M. The Role of TNF-α in Neuropathic Pain: An Immunotherapeutic Perspective. Life 2025, 15, 785. [Google Scholar] [CrossRef] [PubMed]
  168. Hameed, S. Nav1.7 and Nav1.8: Role in the pathophysiology of pain. Mol. Pain 2019, 15, 1744806919858801. [Google Scholar] [CrossRef]
  169. Pereira, M.; Gazzinelli, R.T. Regulation of innate immune signaling by IRAK proteins. Front. Immunol. 2023, 14, 1133354. [Google Scholar] [CrossRef]
  170. Jeevakumar, V.; Al Sardar, A.K.; Mohamed, F.; Smithhart, C.M.; Price, T.; Dussor, G. IL-6 induced upregulation of T-type Ca2+ currents and sensitization of DRG nociceptors is attenuated by MNK inhibition. J. Neurophysiol. 2020, 124, 274–283. [Google Scholar] [CrossRef]
  171. St-Jacques, B.; Ma, W. Prostaglandin E2/EP4 signalling facilitates EP4 receptor externalization in primary sensory neurons in vitro and in vivo. Pain 2013, 154, 313–323. [Google Scholar] [CrossRef] [PubMed]
  172. Schnizler, K.; Shutov, L.P.; Van Kanegan, M.J.; Merrill, M.A.; Nichols, B.; McKnight, G.S.; Strack, S.; Hell, J.W.; Usachev, Y.M. Protein kinase A anchoring via AKAP150 is essential for TRPV1 modulation by forskolin and prostaglandin E2 in mouse sensory neurons. J. Neurosci. 2008, 28, 4904–4917. [Google Scholar] [CrossRef]
  173. Cadiou, H.; Studer, M.; Jones, N.G.; Smith, E.S.; Ballard, A.; McMahon, S.B.; McNaughton, P.A. Modulation of acid-sensing ion channel activity by nitric oxide. J. Neurosci. 2007, 27, 13251–13260. [Google Scholar] [CrossRef]
  174. Spiers, J.G.; Steinert, J.R. Nitrergic modulation of ion channel function in regulating neuronal excitability. Channels 2021, 15, 666–679. [Google Scholar] [CrossRef]
  175. Yoshimura, N.; Seki, S.; de Groat, W.C. Nitric oxide modulates Ca2+ channels in dorsal root ganglion neurons innervating rat urinary bladder. J. Neurophysiol. 2001, 86, 304–311. [Google Scholar] [CrossRef]
  176. Linley, J.E.; Ooi, L.; Pettinger, L.; Kirton, H.; Boyle, J.P.; Peers, C.; Gamper, N. Reactive oxygen species are second messengers of neurokinin signaling in peripheral sensory neurons. Proc. Natl. Acad. Sci. USA 2012, 109, E1578–E1586. [Google Scholar] [CrossRef]
  177. Meacham, K.; Shepherd, A.; Mohapatra, D.P.; Haroutounian, S. Neuropathic Pain: Central vs. Peripheral Mechanisms. Curr. Pain Headache Rep. 2017, 21, 28. [Google Scholar] [CrossRef]
  178. Zhang, X.; Cao, Y.; Li, L.; Liu, Y.; Zhou, P.; Lai, Y.; Wang, S.; Zuo, Y.; Chen, J.; Chen, C.; et al. Quercetin targets the Ccl4-Ccr5 axis to relieve neuropathic pain after spinal cord injury. APL Bioeng. 2025, 9, 036108. [Google Scholar] [CrossRef]
  179. Festa, B.P.; Siddiqi, F.H.; Jimenez-Sanchez, M.; Rubinsztein, D.C. Microglial cytokines poison neuronal autophagy via CCR5, a druggable target. Autophagy 2024, 20, 949–951. [Google Scholar] [CrossRef] [PubMed]
  180. Valiukas, Z.; Tangalakis, K.; Apostolopoulos, V.; Feehan, J. Microglial activation states and their implications for Alzheimer’s Disease. J. Prev. Alzheimers Dis. 2025, 12, 100013. [Google Scholar] [CrossRef] [PubMed]
  181. Smith, J.A.; Das, A.; Ray, S.K.; Banik, N.L. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res. Bull. 2012, 87, 10–20. [Google Scholar] [CrossRef] [PubMed]
  182. Kim, S.; Hahn, Y.K.; Podhaizer, E.M.; McLane, V.D.; Zou, S.; Hauser, K.F.; Knapp, P.E. A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates. J. Neuroinflamm. 2018, 15, 285. [Google Scholar] [CrossRef]
  183. Cappoli, N.; Tabolacci, E.; Aceto, P.; Dello Russo, C. The emerging role of the BDNF-TrkB signaling pathway in the modulation of pain perception. J. Neuroimmunol. 2020, 349, 577406. [Google Scholar] [CrossRef]
  184. Smith, P.A. BDNF in Neuropathic Pain; the Culprit that Cannot be Apprehended. Neuroscience 2024, 543, 49–64. [Google Scholar] [CrossRef] [PubMed]
  185. Breitinger, U.; Breitinger, H.G. Excitatory and inhibitory neuronal signaling in inflammatory and diabetic neuropathic pain. Mol. Med. 2023, 29, 53. [Google Scholar] [CrossRef]
  186. Pittaluga, A. CCL5-Glutamate Cross-Talk in Astrocyte-Neuron Communication in Multiple Sclerosis. Front. Immunol. 2017, 8, 1079. [Google Scholar] [CrossRef] [PubMed]
  187. Ji, R.R.; Befort, K.; Brenner, G.J.; Woolf, C.J. ERK MAP kinase activation in superficial spinal cord neurons induces prodynorphin and NK-1 upregulation and contributes to persistent inflammatory pain hypersensitivity. J. Neurosci. 2002, 22, 478–485. [Google Scholar] [CrossRef]
  188. Lu, Y.; Jiang, B.C.; Cao, D.L.; Zhao, L.X.; Zhang, Y.L. Chemokine CCL8 and its receptor CCR5 in the spinal cord are involved in visceral pain induced by experimental colitis in mice. Brain Res. Bull. 2017, 135, 170–178. [Google Scholar] [CrossRef]
  189. MacDermott, A.B.; Mayer, M.L.; Westbrook, G.L.; Smith, S.J.; Barker, J.L. NMDA-receptor activation increases cytoplasmic calcium concentration in cultured spinal cord neurones. Nature 1986, 321, 519–522. [Google Scholar] [CrossRef]
  190. Simonetti, M.; Hagenston, A.M.; Vardeh, D.; Freitag, H.E.; Mauceri, D.; Lu, J.; Satagopam, V.P.; Schneider, R.; Costigan, M.; Bading, H.; et al. Nuclear calcium signaling in spinal neurons drives a genomic program required for persistent inflammatory pain. Neuron 2013, 77, 43–57. [Google Scholar] [CrossRef] [PubMed]
  191. Musante, V.; Longordo, F.; Neri, E.; Pedrazzi, M.; Kalfas, F.; Severi, P.; Raiteri, M.; Pittaluga, A. RANTES modulates the release of glutamate in human neocortex. J. Neurosci. 2008, 28, 12231–12240. [Google Scholar] [CrossRef]
  192. Sandkühler, J. Understanding LTP in pain pathways. Mol. Pain 2007, 3, 9. [Google Scholar] [CrossRef]
  193. Giri, R.K.; Rajagopal, V.; Shahi, S.; Zlokovic, B.V.; Kalra, V.K. Mechanism of amyloid peptide induced CCR5 expression in monocytes and its inhibition by siRNA for Egr-1. Am. J. Physiol. Cell Physiol. 2005, 289, C264–C276. [Google Scholar] [CrossRef]
  194. Kim, H.K.; Park, H.R.; Sul, K.H.; Chung, H.Y.; Chung, J. Induction of RANTES and CCR5 through NF-κB activation via MAPK pathway in aged rat gingival tissues. Biotechnol. Lett. 2006, 28, 17–23. [Google Scholar] [CrossRef]
  195. Garland, E.F.; Hartnell, I.J.; Boche, D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front. Neurosci. 2022, 16, 824888. [Google Scholar] [CrossRef]
  196. Karavis, M.Y.; Siafaka, I.; Vadalouca, A.; Georgoudis, G. Role of Microglia in Neuropathic Pain. Cureus 2023, 15, e43555. [Google Scholar] [CrossRef] [PubMed]
  197. Loyd, D.R.; Murphy, A.Z. The role of the periaqueductal gray in the modulation of pain in males and females: Are the anatomy and physiology really that different? Neural Plast. 2009, 2009, 462879. [Google Scholar] [CrossRef]
  198. Nimura, F.; Zhang, L.F.; Okuma, K.; Tanaka, R.; Sunakawa, H.; Yamamoto, N.; Tanaka, Y. Cross-linking cell surface chemokine receptors leads to isolation, activation, and differentiation of monocytes into potent dendritic cells. Exp. Biol. Med. 2006, 231, 431–443. [Google Scholar] [CrossRef] [PubMed]
  199. He, J.; Chen, Y.; Farzan, M.; Choe, H.; Ohagen, A.; Gartner, S.; Busciglio, J.; Yang, X.; Hofmann, W.; Newman, W.; et al. CCR3 and CCR5 are co-receptors for HIV-1 infection of microglia. Nature 1997, 385, 645–649. [Google Scholar] [CrossRef] [PubMed]
  200. Dansereau, M.A.; Midavaine, É.; Bégin-Lavallée, V.; Belkouch, M.; Beaudet, N.; Longpré, J.M.; Mélik-Parsadaniantz, S.; Sarret, P. Mechanistic insights into the role of the chemokine CCL2/CCR2 axis in dorsal root ganglia to peripheral inflammation and pain hypersensitivity. J. Neuroinflamm. 2021, 18, 79. [Google Scholar] [CrossRef]
  201. Zhang, Z.J.; Dong, Y.L.; Lu, Y.; Cao, S.; Zhao, Z.Q.; Gao, Y.J. Chemokine CCL2 and its receptor CCR2 in the medullary dorsal horn are involved in trigeminal neuropathic pain. J. Neuroinflamm. 2012, 9, 136. [Google Scholar] [CrossRef]
  202. Fantuzzi, L.; Tagliamonte, M.; Gauzzi, M.C.; Lopalco, L. Dual CCR5/CCR2 targeting: Opportunities for the cure of complex disorders. Cell. Mol. Life Sci. 2019, 76, 4869–4886. [Google Scholar] [CrossRef]
  203. Hwang, C.J.; Park, M.H.; Hwang, J.Y.; Kim, J.H.; Yun, N.Y.; Oh, S.Y.; Song, J.K.; Seo, H.O.; Kim, Y.B.; Hwang, D.Y.; et al. CCR5 deficiency accelerates lipopolysaccharide-induced astrogliosis, amyloid-beta deposit and impaired memory function. Oncotarget 2016, 7, 11984–11999. [Google Scholar] [CrossRef]
  204. Bober, A.; Mika, J.; Piotrowska, A. A Missing Puzzle in Preclinical Studies—Are CCR2, CCR5, and Their Ligands’ Roles Similar in Obesity-Induced Hypersensitivity and Diabetic Neuropathy?—Evidence from Rodent Models and Clinical Studies. Int. J. Mol. Sci. 2024, 25, 11323. [Google Scholar] [CrossRef]
  205. Bedognetti, D.; Spivey, T.L.; Zhao, Y.; Uccellini, L.; Tomei, S.; Dudley, M.E.; Ascierto, M.L.; De Giorgi, V.; Liu, Q.; Delogu, L.G.; et al. CXCR3/CCR5 pathways in metastatic melanoma patients treated with adoptive therapy and interleukin-2. Br. J. Cancer 2013, 109, 2412–2423. [Google Scholar] [CrossRef] [PubMed]
  206. Ni, J.; Zhu, Y.N.; Zhong, X.G.; Ding, Y.; Hou, L.F.; Tong, X.K.; Tang, W.; Ono, S.; Yang, Y.F.; Zuo, J.P. The chemokine receptor antagonist, TAK-779, decreased experimental autoimmune encephalomyelitis by reducing inflammatory cell migration into the central nervous system, without affecting T cell function. Br. J. Pharmacol. 2009, 158, 2046–2056. [Google Scholar] [CrossRef]
  207. Gao, Y.J.; Ji, R.R. Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol. Ther. 2010, 126, 56–68. [Google Scholar] [CrossRef]
  208. Kwiatkowski, K.; Piotrowska, A.; Rojewska, E.; Makuch, W.; Jurga, A.; Slusarczyk, J.; Trojan, E.; Basta-Kaim, A.; Mika, J. Beneficial properties of maraviroc on neuropathic pain development and opioid effectiveness in rats. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2016, 64, 68–78. [Google Scholar] [CrossRef] [PubMed]
  209. Kwiatkowski, K.; Ciapała, K.; Rojewska, E.; Makuch, W.; Mika, J. Comparison of the beneficial effects of RS504393, maraviroc and cenicriviroc on neuropathic pain-related symptoms in rodents: Behavioral and biochemical analyses. Int. Immunopharmacol. 2020, 84, 106540. [Google Scholar] [CrossRef]
  210. Ciechanowska, A.; Pawlik, K.; Ciapała, K.; Mika, J. Pharmacological Modulation of the MIP-1 Family and Their Receptors Reduces Neuropathic Pain Symptoms and Influences Morphine Analgesia: Evidence from a Mouse Model. Brain Sci. 2023, 13, 579. [Google Scholar] [CrossRef] [PubMed]
  211. Padi, S.S.V.; Shi, X.Q.; Zhao, Y.Q.; Ruff, M.R.; Baichoo, N.; Pert, C.B.; Zhang, J. Attenuation of rodent neuropathic pain by an orally active peptide, RAP-103, which potently blocks CCR2- and CCR5-mediated monocyte chemotaxis and inflammation. Pain 2012, 153, 95–106. [Google Scholar] [CrossRef]
  212. Saika, F.; Kiguchi, N.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. CC-chemokine ligand 4/macrophage inflammatory protein-1β participates in the induction of neuropathic pain after peripheral nerve injury. Eur. J. Pain 2012, 16, 1271–1280. [Google Scholar] [CrossRef] [PubMed]
  213. Zhong, S.; Liu, F.; Giniatullin, R.; Jolkkonen, J.; Li, Y.; Zhou, Z.; Lin, X.; Liu, C.; Zhang, X.; Liu, Z.; et al. Blockade of CCR5 suppresses paclitaxel-induced peripheral neuropathic pain caused by increased deoxycholic acid. Cell Rep. 2023, 42, 113386. [Google Scholar] [CrossRef] [PubMed]
  214. Woollard, S.M.; Kanmogne, G.D. Maraviroc: A review of its use in HIV infection and beyond. Drug Des. Dev. Ther. 2015, 9, 5447–5468. [Google Scholar] [CrossRef]
  215. Ray, N. Maraviroc in the treatment of HIV infection. Drug Des. Dev. Ther. 2009, 2, 151–161. [Google Scholar] [CrossRef]
  216. Rockstroh, J.K.; Plonski, F.; Bansal, M.; Fätkenheuer, G.; Small, C.B.; Asmuth, D.M.; Pialoux, G.; Zhang-Roper, R.; Wang, R.; Pineda, J.A.; et al. Hepatic safety of maraviroc in patients with HIV-1 and hepatitis C and/or B virus: 144-week results from a randomized, placebo-controlled trial. Antivir. Ther. 2017, 22, 263–269. [Google Scholar] [CrossRef]
  217. Lefebvre, E.; Gottwald, M.; Lasseter, K.; Chang, W.; Willett, M.; Smith, P.F.; Somasunderam, A.; Utay, N.S. Pharmacokinetics, Safety, and CCR2/CCR5 Antagonist Activity of Cenicriviroc in Participants with Mild or Moderate Hepatic Impairment. Clin. Transl. Sci. 2016, 9, 139–148. [Google Scholar] [CrossRef]
  218. Friedman, S.; Sanyal, A.; Goodman, Z.; Lefebvre, E.; Gottwald, M.; Fischer, L.; Ratziu, V. Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR Phase 2b study design. Contemp. Clin. Trials 2016, 47, 356–365. [Google Scholar] [CrossRef]
  219. Anstee, Q.M.; Neuschwander-Tetri, B.A.; Wai-Sun Wong, V.; Abdelmalek, M.F.; Rodriguez-Araujo, G.; Landgren, H.; Park, G.S.; Bedossa, P.; Alkhouri, N.; Tacke, F.; et al. Cenicriviroc Lacked Efficacy to Treat Liver Fibrosis in Nonalcoholic Steatohepatitis: AURORA Phase III Randomized Study. Clin. Gastroenterol. Hepatol. 2024, 22, 124–134.e1. [Google Scholar] [CrossRef]
  220. Takashima, K.; Miyake, H.; Kanzaki, N.; Tagawa, Y.; Wang, X.; Sugihara, Y.; Iizawa, Y.; Baba, M. Highly potent inhibition of human immunodeficiency virus type 1 replication by TAK-220, an orally bioavailable small-molecule CCR5 antagonist. Antimicrob. Agents Chemother. 2005, 49, 3474–3482. [Google Scholar] [CrossRef] [PubMed]
  221. Gerlag, D.M.; Hollis, S.; Layton, M.; Vencovský, J.; Szekanecz, Z.; Braddock, M.; Tak, P.P.; ESCAPE Study Group. Preclinical and clinical investigation of a CCR5 antagonist, AZD5672, in patients with rheumatoid arthritis receiving methotrexate. Arthritis Rheum. 2010, 62, 3154–3160. [Google Scholar] [CrossRef]
  222. van Kuijk, A.W.R.; Vergunst, C.E.; Gerlag, D.M.; Bresnihan, B.; Gomez-Reino, J.J.; Rouzier, R.; Verschueren, P.C.; van de Leij, C.; Maas, M.; Kraan, M.C.; et al. CCR5 blockade in rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Ann. Rheum. Dis. 2010, 69, 2013–2016. [Google Scholar] [CrossRef]
  223. Noda, M.; Tomonaga, D.; Kitazono, K.; Yoshioka, Y.; Liu, J.; Rousseau, J.P.; Kinkead, R.; Ruff, M.R.; Pert, C.B. Neuropathic pain inhibitor, RAP-103, is a potent inhibitor of microglial CCL1/CCR8. Neurochem. Int. 2018, 119, 184–189. [Google Scholar] [CrossRef] [PubMed]
  224. Alghibiwi, H.; Ansari, M.A.; Nadeem, A.; Algonaiah, M.A.; Attia, S.M.; Bakheet, S.A.; Albekairi, T.H.; Almudimeegh, S.; Alhamed, A.S.; Shahid, M.; et al. DAPTA, a C-C Chemokine Receptor 5 (CCR5), Leads to the Downregulation of Notch/NF-κB Signaling and Proinflammatory Mediators in CD40+ Cells in Experimental Autoimmune Encephalomyelitis Model in SJL/J Mice. Biomedicines 2023, 11, 1511. [Google Scholar] [CrossRef] [PubMed]
  225. Ruff, M.R.; Polianova, M.; Yang, Q.E.; Leoung, G.S.; Ruscetti, F.W.; Pert, C.B. Update on D-ala-peptide T-amide (DAPTA): A viral entry inhibitor that blocks CCR5 chemokine receptors. Curr. HIV Res. 2003, 1, 51–67. [Google Scholar] [CrossRef]
  226. Arribas López, J.R. Efectos secundarios asociados al tratamiento con maraviroc y otros antagonistas de CCR5. Impacto potencial del bloqueo del receptor CCR5. Enferm. Infecc. Microbiol. Clin. 2008, 26, 23–27. [Google Scholar] [CrossRef] [PubMed]
  227. Crespo, M.; Navarro, J.; Moreno, S.; Sanz, J.; Márquez, M.; Zamora, J.; Ocampo, A.; Iribaren, J.A.; Rivero, A.; Llibre, J.M. Hepatic safety of maraviroc in HIV-1-infected patients with hepatitis C and/or B co-infection. The Maraviroc Cohort Spanish Group. Enferm. Infecc. Microbiol. Clin. 2017, 35, 493–498. [Google Scholar] [CrossRef]
  228. Francque, S.M.; Hodge, A.; Boursier, J.; Younes, Z.H.; Rodriguez-Araujo, G.; Park, G.S.; Alkhouri, N.; Abdelmalek, M.F. Phase 2, open-label, rollover study of cenicriviroc for liver fibrosis associated with metabolic dysfunction-associated steatohepatitis. Hepatol. Commun. 2024, 8, e0335. [Google Scholar] [CrossRef]
  229. Abel, S.; Russell, D.; Whitlock, L.A.; Ridgway, C.E.; Nedderman, A.N.; Walker, D.K. Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects. Br. J. Clin. Pharmacol. 2008, 65 (Suppl. S1), 60–67. [Google Scholar] [CrossRef]
  230. Wu, Q.L.; Cui, L.Y.; Ma, W.Y.; Wang, S.S.; Zhang, Z.; Feng, Z.P.; Sun, H.S.; Chu, S.F.; He, W.B.; Chen, N.H. A novel small-molecular CCR5 antagonist promotes neural repair after stroke. Acta Pharmacol. Sin. 2023, 44, 1935–1947. [Google Scholar] [CrossRef]
  231. Keizer, R.J.; Huitema, A.D.; Schellens, J.H.; Beijnen, J.H. Clinical pharmacokinetics of therapeutic monoclonal antibodies. Clin. Pharmacokinet. 2010, 49, 493–507. [Google Scholar] [CrossRef]
  232. Gathe, J.C.; Dejesus, E.; Ramgopal, M.N.; Rolle, C.P.; Yang, O.O.; Sanchez, W.E.; Lalezari, J.P.; Krishen, A.; Sacha, J.B.; Hansen, S.G.; et al. Leronlimab Treatment for Multidrug-Resistant HIV-1 (OPTIMIZE): A Randomized, Double-Blind, Placebo-Controlled Trial. J. Acquir. Immune Defic. Syndr. 2025, 99, 185–194. [Google Scholar] [CrossRef] [PubMed]
  233. Jiao, X.; Wang, M.; Zhang, Z.; Li, Z.; Ni, D.; Ashton, A.W.; Tang, H.Y.; Speicher, D.W.; Pestell, R.G. Leronlimab, a humanized monoclonal antibody to CCR5, blocks breast cancer cellular metastasis and enhances cell death induced by DNA damaging chemotherapy. Breast Cancer Res. 2021, 23, 11. [Google Scholar] [CrossRef]
  234. Yang, B.; Fulcher, J.A.; Ahn, J.; Berro, M.; Goodman-Meza, D.; Dhody, K.; Sacha, J.B.; Naeim, A.; Yang, O.O. Clinical Characteristics and Outcomes of Coronavirus Disease 2019 Patients Who Received Compassionate-Use Leronlimab. Clin. Infect. Dis. 2021, 73, e4082–e4089. [Google Scholar] [CrossRef] [PubMed]
  235. Sánchez-Robles, E.M.; Girón, R.; Paniagua, N.; Rodríguez-Rivera, C.; Pascual, D.; Goicoechea, C. Monoclonal Antibodies for Chronic Pain Treatment: Present and Future. Int. J. Mol. Sci. 2021, 22, 10325. [Google Scholar] [CrossRef]
  236. Nwagwu, C.D.; Sarris, C.; Tao, Y.X.; Mammis, A. Biomarkers for Chronic Neuropathic Pain and their Potential Application in Spinal Cord Stimulation: A Review. Transl. Perioper. Pain Med. 2016, 1, 33–38. [Google Scholar]
  237. Yukselten, Y.; Wishah, H.; Li, J.A.; Sutton, R.E. Targeting CCR5: A central approach to HIV treatment and cure strategies. Virology 2025, 603, 110375. [Google Scholar] [CrossRef] [PubMed]
  238. Akgün, E.; Javed, M.I.; Lunzer, M.M.; Powers, M.D.; Sham, Y.Y.; Watanabe, Y.; Portoghese, P.S. Inhibition of Inflammatory and Neuropathic Pain by Targeting a Mu Opioid Receptor/Chemokine Receptor5 Heteromer (MOR-CCR5). J. Med. Chem. 2015, 58, 8647–8657. [Google Scholar] [CrossRef]
  239. Kim, M.B.; Giesler, K.E.; Tahirovic, Y.A.; Truax, V.M.; Liotta, D.C.; Wilson, L.J. CCR5 receptor antagonists in preclinical to phase II clinical development for treatment of HIV. Expert Opin. Investig. Drugs 2016, 25, 1377–1392. [Google Scholar] [CrossRef]
  240. Wang, J.; Qu, C.; Xiao, P.; Liu, S.; Sun, J.P.; Ping, Y.Q. Progress in structure-based drug development targeting chemokine receptors. Front. Pharmacol. 2025, 16, 1603950. [Google Scholar] [CrossRef]
  241. Zhang, F.; Yuan, Y.; Xiang, M.; Guo, Y.; Li, M.; Liu, Y.; Pu, X. Molecular Mechanism Regarding Allosteric Modulation of Ligand Binding and the Impact of Mutations on Dimerization for CCR5 Homodimer. J. Chem. Inf. Model. 2019, 59, 1965–1976. [Google Scholar] [CrossRef] [PubMed]
  242. Olson, W.C.; Jacobson, J.M. CCR5 monoclonal antibodies for HIV-1 therapy. Curr. Opin. HIV AIDS 2009, 4, 104–111. [Google Scholar] [CrossRef] [PubMed]
  243. Saita, Y.; Kondo, M.; Miyazaki, T.; Yamaji, N.; Shimizu, Y. Transgenic mouse expressing human CCR5 as a model for in vivo assessments of human selective CCR5 antagonists. Eur. J. Pharmacol. 2005, 518, 227–233. [Google Scholar] [CrossRef] [PubMed]
  244. Zhou, Z.; Cong, L.; Cong, X. Patient-Derived Organoids in Precision Medicine: Drug Screening, Organoid-on-a-Chip and Living Organoid Biobank. Front. Oncol. 2021, 11, 762184. [Google Scholar] [CrossRef]
  245. Ellinger, B.; Pohlmann, D.; Woens, J.; Jäkel, F.M.; Reinshagen, J.; Stocking, C.; Prassolov, V.S.; Fehse, B.; Riecken, K. A High-Throughput HIV-1 Drug Screening Platform, Based on Lentiviral Vectors and Compatible with Biosafety Level-1. Viruses 2020, 12, 580. [Google Scholar] [CrossRef]
  246. Staudt, M.D. The Multidisciplinary Team in Pain Management. Neurosurg. Clin. N. Am. 2022, 33, 241–249. [Google Scholar] [CrossRef]
Figure 1. Intracellular signaling pathways mediated by CCR5. Binding of CCR5 ligands (principally CCL3, CCL4, and CCL5) to the CCR5 receptor activates several heterotrimeric G proteins, whose distinct subunits subsequently initiate specific signaling cascades: (1) Gαq stimulates PLC-β, leading to the activation of CaMK and PKC; (2) Gαi inhibits adenylate cyclase, thereby reducing intracellular cAMP levels; (3) the Gβγ subunit activates PI3K-γ/β, which subsequently triggers PKB and the mTOR pathway. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCL3 (chemokine -C-C motif- ligand 3), CCL4 (chemokine -C-C motif- ligand 4), CCL5 (chemokine -C-C motif- ligand 5), Gαq (G protein alpha q subunit), Gαi (G protein alpha i subunit), PLC-β (phospholipase C beta), CaMK (Ca2+/calmodulin-dependent protein kinase), PKC (protein kinase C), cAMP (cyclic adenosine monophosphate), PI3K-γ (phosphatidylinositol 3-kinase gamma), PI3K-β (phosphatidylinositol 3-kinase beta), PKB (protein kinase B), and mTOR (mammalian target of rapamycin).
Figure 1. Intracellular signaling pathways mediated by CCR5. Binding of CCR5 ligands (principally CCL3, CCL4, and CCL5) to the CCR5 receptor activates several heterotrimeric G proteins, whose distinct subunits subsequently initiate specific signaling cascades: (1) Gαq stimulates PLC-β, leading to the activation of CaMK and PKC; (2) Gαi inhibits adenylate cyclase, thereby reducing intracellular cAMP levels; (3) the Gβγ subunit activates PI3K-γ/β, which subsequently triggers PKB and the mTOR pathway. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCL3 (chemokine -C-C motif- ligand 3), CCL4 (chemokine -C-C motif- ligand 4), CCL5 (chemokine -C-C motif- ligand 5), Gαq (G protein alpha q subunit), Gαi (G protein alpha i subunit), PLC-β (phospholipase C beta), CaMK (Ca2+/calmodulin-dependent protein kinase), PKC (protein kinase C), cAMP (cyclic adenosine monophosphate), PI3K-γ (phosphatidylinositol 3-kinase gamma), PI3K-β (phosphatidylinositol 3-kinase beta), PKB (protein kinase B), and mTOR (mammalian target of rapamycin).
Biomedicines 13 02650 g001
Figure 2. CCR5-mediated microglial modulation of spinal projection neurons in pain signaling. Activation of CCR5 in microglia leads to the release of BDNF. BDNF acts on TrkB receptors in spinal projection neurons, reducing KCC-mediated Cl extrusion. This results in increased intracellular Cl and impaired inhibitory neurotransmission through GABA and glycine receptors. Consequently, excitatory glutamatergic transmission is enhanced, promoting pain signaling. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCL3 (chemokine -C-C motif- ligand 3), CCL4 (chemokine -C-C motif- ligand 4), CCL5 (chemokine -C-C motif- ligand 5), BDNF (brain-derived neurotrophic factor), TrkB (tropomyosin receptor kinase B), KCC (K+-Cl cotransporter), [Cli] (intracellular Cl concentration), and GABA (gamma-aminobutyric acid).
Figure 2. CCR5-mediated microglial modulation of spinal projection neurons in pain signaling. Activation of CCR5 in microglia leads to the release of BDNF. BDNF acts on TrkB receptors in spinal projection neurons, reducing KCC-mediated Cl extrusion. This results in increased intracellular Cl and impaired inhibitory neurotransmission through GABA and glycine receptors. Consequently, excitatory glutamatergic transmission is enhanced, promoting pain signaling. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCL3 (chemokine -C-C motif- ligand 3), CCL4 (chemokine -C-C motif- ligand 4), CCL5 (chemokine -C-C motif- ligand 5), BDNF (brain-derived neurotrophic factor), TrkB (tropomyosin receptor kinase B), KCC (K+-Cl cotransporter), [Cli] (intracellular Cl concentration), and GABA (gamma-aminobutyric acid).
Biomedicines 13 02650 g002
Table 1. CCR5 antagonists in preclinical neuropathic pain models. Abbreviations: CCI (chronic constriction injury), CCR5 (C-C chemokine receptor type 5), i.t. (intrathecal injection), p38 MAPK (p38 mitogen-activated protein kinase), ERK1/2 (extracellular signal-regulated kinases 1 and 2), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), STAT3 (signal transducer and activator of transcription 3), DRG (dorsal root ganglion), IL-1β (interleukin 1 beta), IL-18 (interleukin 18), IL-6 (interleukin 6), NOS2 (nitric oxide synthase 2), IL-1RA (Interleukin 1 receptor antagonist), IL-18BP (Interleukin 18 binding protein), IL-10 (Interleukin 10), Iba-1 (ionized calcium-binding adapter molecule 1), GFAP (glial fibrillary acidic protein), qRT-PCR (quantitative real-time polymerase chain reaction), CCL3 (chemokine (C-C motif) ligand 3), CCL4 (chemokine (C-C motif) ligand 4), CCL5 (chemokine (C-C motif) ligand 5), CCL7 (chemokine (C-C motif) ligand 7), DPN (diabetic peripheral neuropathy), mRNA (messenger RNA), STZ (streptozocin), CCL8 (chemokine (C-C motif) ligand 8), CCL12 (chemokine (C-C motif) ligand 12), PSNL (partial sciatic nerve ligation), DAPTA (D-Ala1-peptide T-amide), and PIPN (paclitaxel-induced peripheral neuropathy).
Table 1. CCR5 antagonists in preclinical neuropathic pain models. Abbreviations: CCI (chronic constriction injury), CCR5 (C-C chemokine receptor type 5), i.t. (intrathecal injection), p38 MAPK (p38 mitogen-activated protein kinase), ERK1/2 (extracellular signal-regulated kinases 1 and 2), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), STAT3 (signal transducer and activator of transcription 3), DRG (dorsal root ganglion), IL-1β (interleukin 1 beta), IL-18 (interleukin 18), IL-6 (interleukin 6), NOS2 (nitric oxide synthase 2), IL-1RA (Interleukin 1 receptor antagonist), IL-18BP (Interleukin 18 binding protein), IL-10 (Interleukin 10), Iba-1 (ionized calcium-binding adapter molecule 1), GFAP (glial fibrillary acidic protein), qRT-PCR (quantitative real-time polymerase chain reaction), CCL3 (chemokine (C-C motif) ligand 3), CCL4 (chemokine (C-C motif) ligand 4), CCL5 (chemokine (C-C motif) ligand 5), CCL7 (chemokine (C-C motif) ligand 7), DPN (diabetic peripheral neuropathy), mRNA (messenger RNA), STZ (streptozocin), CCL8 (chemokine (C-C motif) ligand 8), CCL12 (chemokine (C-C motif) ligand 12), PSNL (partial sciatic nerve ligation), DAPTA (D-Ala1-peptide T-amide), and PIPN (paclitaxel-induced peripheral neuropathy).
Neuropathic
Pain Model
CompoundObserved OutcomesProposed MechanismsReferences
CCIMaravirocChronic i.t. administration of maraviroc attenuated neuropathic pain symptoms and elevated the nociceptive threshold approximately 60 min post-administration on days 3 and 7 following CCIMaraviroc suppressed phosphorylated p38 MAPK, ERK1/2, and NF-κB expression while enhancing STAT3 in the spinal cord and DRG
Maraviroc reduced classical pro-nociceptive markers (IL-1β, IL-18, IL-6, and NOS2) and upregulated anti-nociceptive markers (IL-1RA, IL-18BP, and IL-10) in the spinal cord
[54]
CCIMaravirocChronic i.t. administration of maraviroc alleviated neuropathic pain symptoms on day 7 post-CCIMaraviroc decreased Iba-1 and GFAP protein levels and restored CCR5 expression altered by CCI in the spinal cord and DRG
qRT-PCR showed that CCR5 and its pro-nociceptive ligands (CCL3, CCL4, and CCL5) were upregulated after nerve injury, while maraviroc attenuated these increases
[208]
CCIMaraviroc
Cenicriviroc
Reduction in neuropathic
pain outcomes
Maraviroc reduced CCI-induced increases in CCL4 within the spinal cord and selectively decreased CCL5 expression in the DRG
In contrast, cenicriviroc exerted a broader effect, reducing CCL2, CCL3, CCL4, and CCL7 in the spinal cord, and CCL2, CCL3, CCL4, CCL5, and CCL7 in the DRG
[209]
DPNCenicrivirocA single dose of cenicriviroc produced comparable analgesia in male and female mice
Repeated cenicriviroc elicited the most robust and sustained antinociceptive effect, reducing STZ-induced hypersensitivity without tolerance
In male mice, mRNA levels of CCL2, CCL5, and CCL7 were upregulated, whereas female mice exhibited additional upregulation of CCL8 and CCL12[55]
PSNLMaraviroci.t. administration of maraviroc following PSL significantly attenuated mechanical allodynia exclusively in male miceNot defined[47]
CCITAK-220
AZD-5672
Single intrathecal administration of TAK-220 and AZD-5672 dose-dependently reduced pain-related behaviors following CCINot defined[210]
CCIRAP-103Oral administration of RAP-103 robustly attenuated nerve injury-induced mechanical and thermal hypersensitivityRAP-103 treatment slightly reduced GFAP expression, while Iba-1-positive cells in the spinal cord exhibited smaller cell bodies, elongated fine processes, and markedly reduced Iba-1 staining[211]
PSNLDAPTA
(Adaptavir)
DAPTA administration prevented the development of tactile allodynia and thermal hyperalgesiaNot defined[212]
PIPNMaravirocAdministration of the CCR5 antagonist maraviroc attenuates the development of neuropathic pain-related nociceptive behaviorsNot defined[213]
Table 2. Comparative pharmacological characterization of CCR5 and dual CCR2/CCR5 antagonists discussed above. Abbreviations: CCR5 (C-C chemokine receptor type 5), HIV-1 (human immunodeficiency virus type 1), FDA (Food and Drug Administration), CCR2 (C-C chemokine receptor type 2), MASH (metabolic dysfunction-associated steatohepatitis), and PK (pharmacokinetics).
Table 2. Comparative pharmacological characterization of CCR5 and dual CCR2/CCR5 antagonists discussed above. Abbreviations: CCR5 (C-C chemokine receptor type 5), HIV-1 (human immunodeficiency virus type 1), FDA (Food and Drug Administration), CCR2 (C-C chemokine receptor type 2), MASH (metabolic dysfunction-associated steatohepatitis), and PK (pharmacokinetics).
CompoundTargetIndicationsMechanisms
of Action
PharmacokineticsClinical
Status
References
MaravirocCCR5
antagonist
HIV-1Blocks CCR5 to inhibit
HIV-1 entry and immune
cell trafficking
Oral administration
t½ 14–18 h
Activation of CYP3A4
FDA-
approved
[214,215,216]
CenicrivirocCCR2/CCR5
antagonist
MASH
Fibrosis
HIV-1
Blocks CCR2 and CCR5-
associated inflammatory
pathways
Oral administration
t½ 22–42 h
Poor CYP activation
Phase 3
discontinued
[217,218,219]
TAK-220CCR5
antagonist
HIV-1Competitive
CCR5 inhibitor
Good oral
bioavailability
Not tested[220]
AZD-5672CCR5
antagonist
Autoimmune
diseases
Blocks CCR5-mediated
T-cell trafficking
Oral administration
Limited PK data
Phase 2[221,222]
RAP-103CCR2/CCR5
antagonist
Severe psoriasisReduces macrophage
activation
Oral administration
Limited PK data
Phase 2[223]
[NCT07204639]
DAPTA
(Adaptavir)
CCR5
antagonist
Neuroinflammation
HIV-1
Blocks CCR5 and
reduces microglial
activation
Intranasal or
Injectable drug
delivery
Not tested[224,225]
Table 3. Adverse effect profile of CCR5 antagonists. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCR2 (C-C chemokine receptor type 2), ALT (alanine aminotransferase), and AST (aspartate aminotransferase).
Table 3. Adverse effect profile of CCR5 antagonists. Abbreviations: CCR5 (C-C chemokine receptor type 5), CCR2 (C-C chemokine receptor type 2), ALT (alanine aminotransferase), and AST (aspartate aminotransferase).
CompoundTargetSide EffectsReferences
MaravirocCCR5
antagonist
Gastrointestinal symptoms: diarrhea, nausea, and hepatotoxicity
Neurological symptoms: headache, dizziness and insomnia
Respiratory symptoms: cough
Musculoskeletal issues: muscle spasms and back pain
Dermatological problems: rash
General symptoms: weakness
[226,227]
CenicrivirocCCR2/CCR5
antagonist
Gastrointestinal symptoms: diarrhea, nausea, constipation, and abdominal pain
Respiratory symptoms: nasopharyngitis and influenza
Dermatological problems: rash
General symptoms: fatigue, arthralgia, and headache
[228]
AZD-5672CCR5
antagonist
Gastrointestinal symptoms: nausea, diarrhea, abdominal pain, and dyspepsia
Respiratory issues: nasopharyngitis, sinusitis, and bronchitis
Dermatological problems: rash and pruritus
General symptoms: headache, fatigue, and dizziness
Laboratory changes: transient neutropenia and increases in liver enzymes (ALT/AST)
[221]
RAP-103CCR2/CCR5
antagonist
Not published[NCT07204639]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

García-Domínguez, M. The Role of the CCR5 Receptor in Neuropathic Pain Modulation: Current Insights and Therapeutic Implications. Biomedicines 2025, 13, 2650. https://doi.org/10.3390/biomedicines13112650

AMA Style

García-Domínguez M. The Role of the CCR5 Receptor in Neuropathic Pain Modulation: Current Insights and Therapeutic Implications. Biomedicines. 2025; 13(11):2650. https://doi.org/10.3390/biomedicines13112650

Chicago/Turabian Style

García-Domínguez, Mario. 2025. "The Role of the CCR5 Receptor in Neuropathic Pain Modulation: Current Insights and Therapeutic Implications" Biomedicines 13, no. 11: 2650. https://doi.org/10.3390/biomedicines13112650

APA Style

García-Domínguez, M. (2025). The Role of the CCR5 Receptor in Neuropathic Pain Modulation: Current Insights and Therapeutic Implications. Biomedicines, 13(11), 2650. https://doi.org/10.3390/biomedicines13112650

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop