Next Article in Journal
Application of Orthobiologics in Achilles Tendinopathy: A Review
Next Article in Special Issue
Changes in mGlu5 Receptor Signaling Are Associated with Associative Learning and Memory Extinction in Mice
Previous Article in Journal
Serum Osteopontin Level Is Positively Associated with Aortic Stiffness in Patients with Peritoneal Dialysis
Previous Article in Special Issue
In Silico Screening for Pesticide Candidates against the Desert Locust Schistocerca gregaria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain

by
Martina Vincenzi
1,*,
Michele Stanislaw Milella
2,
Ginevra D’Ottavio
3,4,
Daniele Caprioli
3,4,
Ingrid Reverte
1,3,* and
Daniela Maftei
1,3
1
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
2
Toxicology and Poison Control Center Unit, Department of Emergency, Anesthesia and Critical Care, Policlinico Umberto I Hospital-Sapienza University of Rome, 00161 Rome, Italy
3
Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy
4
Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
*
Authors to whom correspondence should be addressed.
Life 2022, 12(3), 398; https://doi.org/10.3390/life12030398
Submission received: 15 February 2022 / Revised: 6 March 2022 / Accepted: 7 March 2022 / Published: 9 March 2022

Abstract

:
Neuropathic pain (NP) originates from an injury or disease of the somatosensory nervous system. This heterogeneous origin and the possible association with other pathologies make the management of NP a real challenge. To date, there are no satisfactory treatments for this type of chronic pain. Even strong opioids, the gold-standard analgesics for nociceptive and cancer pain, display low efficacy and the paradoxical ability to exacerbate pain sensitivity in NP patients. Mounting evidence suggests that chemokine upregulation may be a common mechanism driving NP pathophysiology and chronic opioid use-related consequences (analgesic tolerance and hyperalgesia). Here, we first review preclinical studies on the role of chemokines and chemokine receptors in the development and maintenance of NP. Second, we examine the change in chemokine expression following chronic opioid use and the crosstalk between chemokine and opioid receptors. Then, we examine the effects of inhibiting specific chemokines or chemokine receptors as a strategy to increase opioid efficacy in NP. We conclude that strong opioids, along with drugs that block specific chemokine/chemokine receptor axis, might be the right compromise for a favorable risk/benefit ratio in NP management.

1. Introduction

Pain caused by a lesion or disease of the somatosensory nervous system is defined by the International Association for the Study of Pain (IASP) as neuropathic pain (NP) [1]. A wide range of etiological factors may contribute to the development of NP at the peripheral (peripheral neuropathic pain, PNP) or central level (central neuropathic pain, CNP) [2]. This heterogeneity and the widespread association with mood and/or sleep disorders [3], persistent inflammatory conditions and comorbidities [4,5,6] may cause underdiagnosing and undertreating neuropathic conditions [2,7]. Nowadays, the treatment of NP is a real challenge for physicians because the pharmacological tools available are not effective in many patients [8,9,10], and the mechanisms underlying the development and maintenance of NP are not fully understood [11,12]. Thus, a structured, stepwise, and multidisciplinary approach is recommended to improve NP diagnosis and treatment [13] (Figure 1).
The current available therapeutic recommendations for NP include both pharmacological (such as antidepressants, antiepileptics or opioids) and non-pharmacological approaches (such as interventional therapies or physical and psychological therapies) [8,9,10,21,22]. In this review, we focused on opioid therapy and refer readers elsewhere for more comprehensive reviews on alternative approaches [9,10,13,21,22].
Opioids, which are commonly prescribed and highly effective for the treatment of moderate to severe pain, are considered only a second or third choice for the treatment of NP [9]. This is because of their abuse potential, analgesia tolerance, and the often-reported exacerbation of pain sensitivity in patients that are already affected by NP [9,27].
Mounting evidence suggests that the mechanisms associated with opioid-induced hyperalgesia (OIH), a state of nociceptive sensitization caused by chronic exposure to opioids, have some commonalities with nerve injury-induced hypersensitivity [27]. It is therefore plausible that the mechanisms that promote OIH and NP might synergize and ultimately exacerbate NP [27,28] in NP patients treated with opioids.
In recent decades, numerous studies have indicated that peripheral nervous system (PNS) or central nervous system (CNS) damage leads to activation of glial cells, which leads to the release of pronociceptive mediators involved in peripheral and subsequent central sensitization [29,30,31,32]. Chemokines and chemokine receptors play a key role in the development and maintenance of the inflammatory components of NP [33,34]. Notably, increased expression of chemokines and chemokine receptors in neuronal and non-neuronal cells has also been described after opioid exposure [35]. The up-regulation of chemokines and the bidirectional interaction between chemokine receptors and opioid receptors (heterologous desensitization and heterodimerization) may therefore directly contribute to OIH and analgesic tolerance, and may explain the reduced efficacy of opioids in NP [36,37,38].
In this review, we first discuss the preclinical literature on the expression of various chemokines and chemokine receptors and their modulation in different animal models of NP. We then examine the changes in chemokine expression following opioid use in conditions with no neuropathic component and the crosstalk between chemokine and opioid receptors. Next, we review preclinical studies on the effects of inhibiting specific chemokines or chemokine receptors and how their blocking may increase opioid efficacy in NP. We conclude that antagonism of specific chemokines and chemokine receptors may not only improve the symptoms of NP per se but may also be clinically useful in increasing the efficacy of opioids in the treatment of NP. Pending the discovery of new and safe analgesics with innovative mechanisms of action, rational polytherapy currently seems to be the right compromise for a favorable risk/benefit ratio in the management of NP.

2. Neuropathological Mechanisms Underlying Neuropathic Pain

NP occurs as a direct consequence of a disease or lesion of the somatosensory nervous system [1]. Depending on which part of the nervous system is primarily affected, NP can be divided into peripheral (due to lesions or diseases of the PNS) and central (due to lesions or diseases of the CNS) [39]. Patients experiencing NP may complain of intermittent or ongoing spontaneous pain, described as burning, shooting, tingling, pricking, pins and needles, or freezing pain, as well as stimulus-evoked types of pain [1,2]. In the last case, exposure to stimuli such as cold or light touch may manifest as either increased sensitivity to the painful stimuli, hyperalgesia, or pain triggered by non-painful stimuli, allodynia [1,2].
So far, the data from preclinical studies (using animal models that mimic the different aspects of human NP—see Table 1; we further refer readers to reviews [40,41]), have shown that NP symptoms are due to diverse anatomical, molecular, and electrophysiological changes which alter the normal sensory signaling from the periphery to the CNS [2].
These changes occur over the course of weeks or months and vary depending on the nature and extent of the disease or injury [104]. However, regardless of the aetiology, NP pathophysiology is characterized by a long-lasting and even permanent sensitization, both peripheral (through an increased stimulation response and a decrease in the nociceptive threshold of afferent nerve fibers [105]) and central (via increased responsiveness of nociceptive neurons in the spinal cord and CNS to their normal or subthreshold afferent inputs [1,106]).
A nerve injury, for instance, may thus increase the sensitization and excitability of primary sensory neurons, enhancing the excitatory synaptic transmission and decreasing the inhibitory transmission in CNS neurons [107,108]. The modulation of excitatory and inhibitory signaling has several neural correlates, such as the following: altered expression of ion channels (Na+, Ca2+, and K+) [109,110,111], up-regulation of certain neurotransmitters and neuromodulators such as substance P, calcitonin gene-related peptide, bradykinin, glutamate and neuropeptide Y, release of adenosine triphosphate, up-regulation of purinergic receptors, changes in protein kinase C and N-methyl-D-aspartate receptor (NMDA) activity, and changes in the expression of growth factors, cytokines, and chemokines [11,12,112,113].
In the last decade, it has been suggested that, in addition to changes in neuronal activity, the activity of non-neuronal cells, represented by immune cells (macrophages and lymphocytes) and glial cells (Schwann cells and satellite cells in the PNS, and microglia and astrocytes in the spinal cord and CNS), plays an important role in the regulation of NP [114,115,116,117,118]. Nerve damage leads, in fact, to the activation of resident macrophages and Schwann cells near the injury site, while satellite cells are activated in the dorsal root ganglia (DRG) [117,119]. Once activated, these cells begin to produce and release various proinflammatory mediators responsible for the recruitment of leukocytes to the site of injury, sensitization of sensory neurons, and activation of spinal glial cells [119]. On the other hand, sensory neurons and activated glial cells may also release proinflammatory factors that further sensitize neurons and lead to peripheral and central sensitization [120]. Activation of immune and glial cells in both the PNS and the CNS contributes to neuroinflammation by producing and releasing proinflammatory cytokines and chemokines, growth factors, and cellular adhesion molecules [31,32,34,121]. Therefore, it is important to understand the sequence and nature of the events underlying neuroimmune communication to find new mechanisms and targets for the treatment of NP.
In this regard, chemokines and chemokine receptors are gaining growing interest as modulators of neuronal plasticity and enhanced nociceptive transmission in NP conditions.
Chemokines are expressed and synthesized by immune and CNS cells under both physiological and pathological conditions [122,123,124]. They act as the key communication molecules between neurons, glial, and immune cells in pathological pain [33,34] and are involved in both peripheral and central sensitization following nerve injury [30,31,32]. Another main role of chemokines is to attract circulating granulocytes, lymphocytes, and monocytes to the site of injury, resulting in an inflammatory response [125]. Of note, it is now known that chemokines and chemokine receptors are expressed not only by immune cells but also by cells of the nervous system (e.g., neurons and glial cells) [122,126,127] (Box 1).
Box 1. Chemokines and chemokine receptors.
Chemokines, or ‘chemotactic cytokines’, are a family of small secreted (8–17 kDa) molecules that can induce directed chemotaxis of immune cells [128,129]. Most chemokines have two names, one referring to their biological activity, such as monocyte chemoattractant protein-1 (MCP-1) and the other to their structure [123]. Based on their structure and the position and number of conserved cysteine residues in the N-terminus, chemokines are classified into the four following subfamilies: CC, CXC, CX3C, and XC [130]. The CC-subfamily is the largest group of chemokines characterized by the adjacent positions of the first two of a total of four cysteine residues [131]. They contribute to a wide range of functions: they attract monocytes, eosinophils, basophils, T lymphocytes, natural killer (NK) cells, and dendritic cells [131,132]. The CXC-subfamily is the second largest group, characterized by a single amino acid separating the two cysteine residues, and is responsible for monocyte and granulocyte migration [133]. The XC-subfamily has only two closely related members (XCL1 and XCL2), characterized by two cysteine residues, and is responsible for the migration of lymphocytes but not neutrophils or monocytes. The CX3X-subfamily has only one member, CX3CL1 (also called fractalkine), which is characterized by three amino acids separating the two cysteine residues and acts as a chemoattractant and adhesion molecule for T lymphocytes, monocytes, and NK cells [119,131,132].
Chemokines exert their functions by activating G protein-coupled receptors (GPCRs) [134]. The nomenclature of chemokine receptors is CCR, CXCR, XCR, or CX3CR, analogous to their ligands. Chemokines within each subclass have promiscuous relationship with their receptors [134]: multiple chemokines can bind to the same receptor and a single chemokine can bind to multiple receptors. The exception to this rule is the interaction between CX3CL1 and its receptor, CX3CR1, which is monogamous [135].
In the case of peripheral nerve damage due to trauma, diseases, or drugs, a large number of chemokines and chemokine receptors are up-regulated in the PNS and/or CNS, and inhibition of these chemokines and chemokine receptors delays or alleviates pain behavior in the corresponding animal models [34,35,124,136]. For example, binding of the chemokine CCL2 (monocyte chemoattractant protein 1, MCP-1), particularly to the CCR2 receptor (C-C chemokine receptor type 2), has been reported to promote neuroinflammation and maintain the NP condition [137,138]. Up-regulation of CCL2 in primary sensory neurons of the DRG [138,139] and in neurons and astrocytes of the spinal cord [137,140] induces strong glial activation through CCR2 binding [141]. In addition, activation of CCR2 leads to activation of the p38MAPK pathway in microglia, resulting in the production of pronociceptive cytokines such as TNFα, IL-1β, IL-6, and IL-18 [30,121,137] and in spinal neurons, contributing to central sensitization through NMDA receptors [30]. Pharmacological or genetic inhibition of CCL2 and/or its receptor CCR2 alleviates pain in several animal models of NP [142,143] and inhibits glial cell activation [144].
CX3CL1 (fractalkine) has also been reported to be involved in the development and maintenance of NP and neuroinflammation [145]. These effects are mediated by the CX3CR1 receptor, whose expression has been shown to increase in spinal microglia after nerve injury [145]. Activation of CX3CR1 by CX3CL1 activates the microglial p38MAPK signaling pathway, leading to the production of pronociceptive molecules, i.e., IL-1β, IL-6, and TNFα [146]. The administration of neutralizing antibodies against CX3CL1 or CX3CR1 delays or attenuates chronic pain-related behaviors and reduces the release of proinflammatory factors [146,147], whereas CX3CR1 knock-out mice exhibit reduced pain behaviors correlated with decreased microglial activity [148].
Although CCL2 and CX3CL1 are the most extensively studied chemokines associated with NP, the roles of other chemokines and their receptors have been investigated in a variety of traumatic and non-traumatic NP conditions [124]. Of these chemokines, CCL1/CCR8, CCL3/CCR1, CCL3/CCR5, CCL4/CCR5, CCL5/CCR5, CCL7/CCR2, CCL21/CCR7, CXCL1/CXCR2, CXCL10/CXCR3, CXCL12/CXCR4, CXCL13/CXCR5, XCL1/XC1 expression has been shown to increase rapidly in primary sensory neurons and satellite cells of the DRG and in neurons and glial cells of the spinal cord, contributing to the development of hyperalgesia and allodynia via glial activation [124]. On the other hand, the inhibition of their functions by specific neutralizing antibodies, specific receptor antagonists, small interfering RNA (siRNA), or genetic ablation, leads to a marked reduction in NP and associated neuroinflammation [34,35,124,136].
Clinical studies analyzing the levels of different chemokines in the body fluids of patients with NP have shown that neuropathy increases the concentration of CX3CL1, CXCL5, CXCL10, CCL8, or CCL11 in cerebrospinal fluid (CSF) [149], CCL2, CCL3, CCL4, CCL19 in plasma [150,151], and CCL3, CCL4 in saliva [151]. Moreover, the high circulating levels of CCL2 revealed in patients with a traumatic spinal cord injury positively correlate with pain intensity [152].
A newly identified chemokine, prokineticin 2 (PK2), has been shown to play a critical role in the immune system and pain [153]. PK2 exerts its effects by activating two G-protein coupled receptors (GPCRs), prokineticin receptor 1 and 2 (PKR1 and 2), which are widely distributed in pain sites such as peripheral nerves, DRG, and the spinal cord [154]. PK2 and its receptors have been shown to be involved in the development and maintenance of experimental NP of various origins [155,156,157]. Indeed, neuropathy increases the expression of PK2 in Schwann cells, satellite cells, and primary sensory neurons of the DRG, astrocytes, and in the presynaptic terminals of the spinal cord [155,157]. On the contrary, blocking PK2 activity via specific PKR antagonists alleviates the neuropathy-induced pain and reduces the neuroinflammatory state in the sensory nerves, DRG, and spinal cord [155,156,157].
Despite the deleterious effects demonstrated for chemokines in NP, there are recent literature studies that suggest a neuroprotective role of some chemokines in other pathological conditions [158,159,160]. For example, the chemokine CCL5 acting on a GPCR named GPR75 (G Protein-coupled Receptor 75), which does not belong to the chemokine receptor family and is expressed by neuronal cells, activates intracellular signaling pathways associated with neuroprotective effects [159]. However, this CCL5/GPR75 neuroprotective effect has not been yet studied in NP conditions.
Considering their wide expression and activity on multiple regulatory pathways, modulation of chemokine/chemokine receptor signaling could represent a valuable target to develop new therapeutic options for NP.

3. Opioid Therapy and the Loss of Strong Opioid Analgesia in Neuropathic Pain

Opioids carry out their analgesic effects by modulating both the descending and ascending pain pathways [161,162], mainly through the activation of μ-opioid receptors (MORs) [163] and are therefore defined as MOR-agonists. Based on their binding affinity for MORs, opioids are classified as either weak or strong opioids. Although strong opioids such as morphine, oxycodone, and fentanyl are the most effective analgesics for the treatment of acute nociceptive pain and cancer pain [164], their usefulness in NP is still controversial due to use-related concerns (tolerance, OIH, abuse) [22,165,166] and the limited efficacy shown in clinical trials [167,168,169,170,171,172,173,174]. Surprisingly, despite the modest affinity for MORs, weak opioids such as tramadol and tapentadol have shown moderate efficacy and safety in various neuropathic conditions [175,176,177,178]. Some authors hypothesized that the better analgesic spectrum of the weak opioids over strong opioids in NP may be due to their dual mechanism of action, MOR-agonism, and noradrenaline/serotonin reuptake inhibition [179,180], whose contribution in antinociception depend on the type of pain treated [181].
Interestingly, the mechanisms behind the low efficacy of strong opioids in NP have not yet been elucidated, although strong opioid analgesia (using morphine as a reference molecule) has been characterized in several animal models of NP. These models have shown reduced drug efficacy in both allodynia [182,183,184,185,186,187,188,189,190,191] and hyperalgesia [182,185,189,190,192,193,194,195] and the development of analgesic tolerance following sustained or repeated opioid exposure. In addition, the administration of strong opioids may worsen rather than relieve NP [196,197,198,199,200]. Morphine treatment, in fact, may intensify the pre-existing allodynia in animals with peripheral injuries [200], and it may prolong allodynia for weeks to months after treatment has ended [196,197,198]. The same exacerbation of nociceptive hypersensitivity achieved with morphine is also achieved with oxycodone and fentanyl, as reported by Green-Fulgham et al. [199].
Some mechanistic explanations have been proposed for the loss of strong opioid effectiveness in neuropathic conditions. Among these, we can report the following: the down-regulation of MOR expression in the spinal cord [184,189] or DRG [188,193], increased methylation of the MOR gene promoter in primary sensory neurons [195], the release of peptides such as dynorphin [182,191] and cholecystokinin-B [186,194], the stimulation of glutamate receptors [195,201], the increase in serotonin levels [187] and a decrease in brain morphine concentration [190]. Moreover, as previously noted by Martinez-Navarro et al. [27], analgesic tolerance and OIH may share some mechanisms with NP, which are responsible for the failure of strong opioids in NP. The need for higher doses of morphine in NP, than in other pain conditions [189,202], and the exacerbation of nociceptive hypersensitivity when strong opioid dosing begins a few days to one month after nerve injury [198,199], support the lower sensitivity of NP to strong opioids and suggest a possible direct relationship between strong opioid analgesia and the type of pain treated. In this framework, neuro-inflammation following neuro-immune activation has been proposed as the common motif for the development and maintenance of opioid tolerance/hyperalgesia and NP [28,203]. Finally, the pro-inflammatory effect of opioids may be related to the up-regulation of cytokines and chemokines on neuronal and non-neuronal cells such as astrocytes, microglia, and immune cells [35,36].

4. Chemokine System as Novel Target for Enhancing Opioid Analgesia in Neuropathic Pain Therapy

A growing body of literature supports a critical role for chemokines in the development of opioid tolerance. For example, CCL2 levels are up-regulated by chronic opioid exposure in the dorsal spinal cord and CSF [204], astrocytes [205], human neurons [206], and human peripheral blood mononuclear cells [207]. In particular, chronic morphine exposure has been shown to enhance CCL2 immunoreactivity in the spinal cord, especially in spinal neurons, which was involved in morphine tolerance development [208,209]. The contribution of spinal neuronal CCL2 via CCR2 signaling to morphine tolerance was demonstrated by injection of a CCL2-neutralizing antibody. This approach resulted in significantly reduced antinociceptive tolerance and spinal microglial activation [208,209]. Moreover, the expression of other chemokines and their receptors, such as CXCL1/CXCR2, CXCL10/CXCR3, and CXCR12/CXCR4, is increased by opioids and seems to be associated with analgesic tolerance [210,211,212,213].
Instead, conflicting results, are available on the involvement of CX3CL1/CX3CR1 in opioid tolerance [214,215,216]. Johnston et al. [215] reported that CX3CL1 can modulate morphine analgesia through the release of interleukin-1 (IL-1) from the dorsal spinal cord. Thus, blocking CX3CR1 with a neutralizing antibody reduced the development of OIH and tolerance while enhancing morphine analgesia [215]. In contrast, Peng et al. [216] found that CX3CL1/CX3CR1 signaling in the spinal cord did not change with chronic morphine exposure, and neither a CX3CL1-neutralizing antibody nor a CX3CR1 antagonist could completely reverse the development of morphine tolerance. According to Peng and colleagues [216], Chen et al. [214] confirmed that the involvement of the CX3CL1/CX3CR1 axis in antinociceptive tolerance may be secondary to the effect of opioid administration on both glial activation and cellular localization of CX3CR1. Opioids may up-regulate the expression of CX3CR1, normally present on microglia, on neurons where CX3CR1 and opioid receptors may interact to form inactive heterodimers [214]. Although further studies are needed to define the role of heterodimers in painful conditions and opioid analgesic tolerance, some authors propose heterodimers as a new pharmacological target to enhance opioid analgesia [217,218,219,220]. For example, the administration of MCC22, a bivalent ligand (a MOR-agonist linked with a spacer to a CCR5-antagonist) of the MOR-CCR5 heteromer, in a mouse model of cisplatin-induced PNP leads to a reduction of hyperalgesia and spinal microglial inflammatory response without tolerance, reward, and alteration of motor function [218]. Comparable results were obtained by Akgün et al. [217] in LPS-treated mice.
In addition to chemokine release and heterodimerization, heterologous desensitization between chemokine- and opioid-receptors may also be involved in opioid tolerance [36]. Both the chemokine and opioid receptors are GPCRs (Box 2), and the activation of one of them by its ligand may promote the COOH-terminal phosphorylation-related inactivation of the other one (present on the same cell), with the loss of the ability to bind G-proteins, and thus activate the signal cascade [36]. No data are instead available on the influence of prokineticin system in opioid analgesia.
Box 2. Focus on GPCRs.
In GPCRs, seven transmembrane domains are linked by alternating intracellular and extracellular loops [221]. In the GPCR-inactive form, the cytoplasmic portion interacts with a heterotrimeric G protein formed by α, β, and ɣ subunits binding the GDP [222]. The agonist bound to the extracellular portion leads to a conformational change in the receptor and to the activation of one or more G-proteins [221,222] through the replacement of GDP with GTP [223]. The result is the dissociation of Gα from Gβɣ subunits [221]. Gβɣ dimer modulates several effectors (enzymes and ion channels), while Gα controls the receptor coupling specificity and the efficacy of Gβɣ modulation of ion channels [224,225,226]. Depending on their amino acid sequences, Gα subunits can be classified in four categories, Gαs, Gαi/o, Gαq/11, and Gα12/13 that differ for the downstream signaling pathway [223]. Generally, the Gαs subfamily activates adenylyl cyclase, whereas the Gαi subfamily inhibits adenylyl cyclase; the Gαq subfamily activates phospholipase C; the Gα12 subfamily is involved in GTP-binding protein regulation [223]. Despite the fact that GPCRs do not share an overall identity in amino acids [227], they are classified into classes A, B, and C based on their sequence homologies [228]. Class A, also named “rhodopsin-like”, is the largest and most studied GPCR subfamily and includes receptors for rhodopsin, biogenic amines, and several peptide ligands. Class B includes receptors for hormones and neuropeptides such as vasoactive intestinal peptide, calcitonin, and glucagon, while class C consists mainly of metabotropic glutamate and γ-amino-butyric acid receptors and calcium receptors [228].
Both chemokine- and opioid-receptors are members of the class A GPCR family [228,229,230,231]. They are typically Gαi/o-coupled receptors [38], but the participation of other G-family members cannot be excluded [232,233,234]. They can activate several signal transduction pathways and lead to diverse responses. For example, both chemokine- and opioid-receptors may directly inhibit the activity of adenylyl cyclase leading to the reduction of intracellular cAMP [235,236]. They can also activate the mitogen-activated protein kinase cascade [38,134], especially the ERK1/2 [237,238] and p38 [239,240] pathways. The phosphoinositide-3-kinase and the following activation of NF-kB have also been described as a signal transduction pathway activated by both chemokines [241,242,243] and opioids [244,245]. Finally, chemokine- and opioid-receptors can stimulate phospholipase C to enhance the production of diacylglycerol and inositol 1,4,5-thriphosphate leading to an increase of the protein kinase C activity and of intracellular calcium levels, respectively [38,134].
Given the involvement of chemokines in the development of both NP and opioid-tolerance/hyperalgesia, several researchers have hypothesized that administration of chemokine-neutralizing antibodies or their receptor antagonists may simultaneously reduce pain-related behaviors and improve opioid efficacy in neuropathic conditions [246,247,248,249,250] (Figure 2).
From the CC-subfamily, CCL1/CCR8, CCL2/CCR2-CCR4, CCL3-CCL4-CCL5/CCR5, CCL3-CCL9/CCR1, CCL7/CCR2, and CCL7-CCL11/CCR3 axes were investigated as novel pharmacological targets in several NP animal models. Noting that CCL1 administration induced mechanical and thermal hypersensitivity in naïve mice and that CCL1 levels in spinal neurons increased 7 days after streptozotocin (STZ) injection, Zychowska et al. [251] demonstrated that CCL1-neutralizing antibody administration reduces pain and improves the efficacy of morphine and buprenorphine in the STZ-diabetic neuropathy model. Similar results were obtained with CCL2- and CCL7-neutralizing antibodies in chronic constriction injury (CCI) mice [252]. The role of the CCL2-CCL7/CCR2 pathway in hypersensitivity and opioid effects has been demonstrated by injecting two different CCR2 antagonists, RS504393 and cenicriviroc, into CCI rats [248,253]. RS504393, a selective CCR2-antagonist, reduced pain-related behaviors and enhanced analgesia of morphine and buprenorphine by increasing mRNA and protein levels of pronociceptive (i.e., IL-1β, IL-18, IL-6, and inducible nitric oxide synthase, iNOS) and antinociceptive (i.e., IL-1α) factors [253], without affecting spinal expression of CCL2 and CCL7 [254]. Instead, cenicriviroc, a dual CCR2/CCR5 antagonist, alleviates mechanical/thermal hypersensitivity in CCI rats by decreasing the expression of CCL2, CCL7, and CCR2 in the spinal cord and CCR5 in the DRG and preventing the up-regulation of various pronociceptive mediators [248,254]. Cenicriviroc also improves the analgesic effect of morphine and buprenorphine, possibly by preventing the down-regulation of opioid receptors induced by the neuropathy in the DRG [248]. In addition, Bogacka et al. [255] reported that CCL2 can exert its pronociceptive effect by binding to CCR4. Thus, blocking CCR4 with its antagonist, C021, may reduce the development of mechanical and thermal hypersensitivity, improve the efficacy of morphine and buprenorphine, and stop the development of morphine tolerance in CCI mice [255].
A blockade of the CCL3-CCL4-CCL5/CCR5 axis with maraviroc, a CCR5 antagonist, also reduces NP symptoms by inhibiting the expression of CCL3, CCL4 and CCL5 and intensifies morphine and buprenorphine analgesia in the CCI neuropathy model [249]. The contribution of CCL3 and CCL9 through CCR1 signaling was investigated in the STZ-diabetic neuropathy model, and in CCI rats [250,256]. The CCL3- and CCL9-neutralizing antibodies and CCR1 antagonist (J113863) alleviated NP, and intensified the analgesic potency of morphine and buprenorphine [250,256]. Recently, the same effects of CCR1 antagonism were obtained by blocking CCR3 with its selective antagonist, SB328437 [257].
From the CXC-subfamily, CXCL10-CXCL11/CXCR3 and CXCR13/CXCR5 are proposed as novel therapeutic strategies to improve the efficacy of opioids in neuropathic diseases [258,259,260,261,262]. Ye et al. [262] found that the CXCL10/CXCR3 axis participates in cancer-induced bone pain (BCP) and that CXCL10, which is transiently up-regulated by morphine administration, can induce acute analgesia. Therefore, the inhibition of CXCL10 with a neutralizing antibody may enhance morphine analgesia [262,263]. CXCL11, another CXCR3 ligand, is up-regulated in spinal neurons and astrocytes during the development of morphine tolerance, and its inhibition by a neutralizing antibody may result in decreased morphine tolerance due to the blockage of the crosstalk between astrocytes and neurons [259]. In addition, Piotrowska et al. [260] suggested that blocking of CXCR3 with a selective antagonist, NBI-74330, could be a potential target to enhance the antinociceptive effect of morphine in CCI rats. Akin to CXCL11, CXCL13, which acts via CXCR5, appears to be involved in morphine tolerance in rats with cancer-induced bone pain [258,261]. Therefore, administration of a CXCR13-neutralizing antibody [261] or small interfering RNA (siRNA) of CXCR5 [258] may enhance morphine analgesia and prevent the development of tolerance. All the above-described novel target strategies to enhance strong opioid efficacy in neuropathic states are summarized in Table 2.

5. Conclusions

Neuropathic pain can be caused by a variety of insults to the peripheral or central somatosensory nervous system, including trauma, inflammation, ischemia, and metabolic or neoplastic disorders. The treatment of NP remains challenging as available analgesics (such as nonsteroidal anti-inflammatory drugs or opioids) fail to relieve pain due to lack of efficacy or serious side effects. The low efficacy of opioids on NP urges the demand for alternative therapeutic strategies. This has encouraged us to review the research on non-neuronal cells and proinflammatory mediators, such as chemokines, as a common mechanism contributing, on one hand, to the pathophysiology of NP, and on the other hand, to the development of opioid-related side effects. We suggest that inhibiting this pathogenic step by blocking chemokine production or chemokine receptor activity may be useful in alleviating the pain condition and increasing the efficacy of opioids. Pending the discovery of new and safe analgesics with innovative mechanisms of action, rational polytherapy currently seems to be the right compromise for a favorable risk/benefit ratio in the management of NP.

Author Contributions

Writing—original draft preparations, M.V. and D.M.; Supervision, D.C.; Writing—Review and Editing, M.V., M.S.M., G.D., D.C., I.R. and D.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. IASP: International Association for the Study of Pain. IASP Taxonomy. Pain Terms. Neuropathic Pain. Available online: https://www.iasp-pain.org/Education/Content.aspx-?ItemNumber=1698 (accessed on 20 June 2021).
  2. Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic pain. Nat. Rev. Dis. Primers 2017, 3, 17002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Attal, N.; Lanteri-Minet, M.; Laurent, B.; Fermanian, J.; Bouhassira, D. The specific disease burden of neuropathic pain: Results of a French nationwide survey. Pain 2011, 152, 2836–2843. [Google Scholar] [CrossRef] [PubMed]
  4. Liedgens, H.; Obradovic, M.; De Courcy, J.; Holbrook, T.; Jakubanis, R. A burden of illness study for neuropathic pain in Europe. Clinicoecon Outcomes Res. 2016, 8, 113–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. McDermott, A.M.; Toelle, T.R.; Rowbotham, D.J.; Schaefer, C.P.; Dukes, E.M. The burden of neuropathic pain: Results from a cross-sectional survey. Eur. J. Pain 2006, 10, 127–135. [Google Scholar] [CrossRef]
  6. O’Connor, A.B. Neuropathic pain: Quality-of-life impact, costs and cost effectiveness of therapy. Pharmacoeconomics 2009, 27, 95–112. [Google Scholar] [CrossRef]
  7. Giovannini, S.; Coraci, D.; Brau, F.; Galluzzo, V.; Loreti, C.; Caliandro, P.; Padua, L.; Maccauro, G.; Biscotti, L.; Bernabei, R. Neuropathic Pain in the Elderly. Diagnostics 2021, 11, 613. [Google Scholar] [CrossRef]
  8. Attal, N.; Bouhassira, D.; Baron, R. Diagnosis and assessment of neuropathic pain through questionnaires. Lancet Neurol. 2018, 17, 456–466. [Google Scholar] [CrossRef]
  9. Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpää, M.; Hansson, P.; Jensen, T.S.; et al. Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet Neurol. 2015, 14, 162–173. [Google Scholar] [CrossRef] [Green Version]
  10. Szok, D.; Tajti, J.; Nyári, A.; Vécsei, L. Therapeutic Approaches for Peripheral and Central Neuropathic Pain. Behav. Neurol. 2019, 2019, 8685954. [Google Scholar] [CrossRef]
  11. Cohen, S.P.; Mao, J. Neuropathic pain: Mechanisms and their clinical implications. BMJ 2014, 348, f7656. [Google Scholar] [CrossRef] [Green Version]
  12. 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] [PubMed]
  13. Bates, D.; Schultheis, B.C.; Hanes, M.C.; Jolly, S.M.; Chakravarthy, K.V.; Deer, T.R.; Levy, R.M.; Hunter, C.W. A Comprehensive Algorithm for Management of Neuropathic Pain. Pain Med. 2019, 20, S2–S12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bennett, M. The LANSS Pain Scale: The Leeds assessment of neuropathic symptoms and signs. Pain 2001, 92, 147–157. [Google Scholar] [CrossRef]
  15. Krause, S.J.; Backonja, M.M. Development of a neuropathic pain questionnaire. Clin. J. Pain 2003, 19, 306–314. [Google Scholar] [CrossRef]
  16. Freynhagen, R.; Baron, R.; Gockel, U.; Tölle, T.R. painDETECT: A new screening questionnaire to identify neuropathic components in patients with back pain. Curr. Med. Res. Opin. 2006, 22, 1911–1920. [Google Scholar] [CrossRef] [PubMed]
  17. Bouhassira, D.; Attal, N.; Alchaar, H.; Boureau, F.; Brochet, B.; Bruxelle, J.; Cunin, G.; Fermanian, J.; Ginies, P.; Grun-Overdyking, A.; et al. Comparison of pain syndromes associated with nervous or somatic lesions and development of a new neuropathic pain diagnostic questionnaire (DN4). Pain 2005, 114, 29–36. [Google Scholar] [CrossRef]
  18. Portenoy, R. Development and testing of a neuropathic pain screening questionnaire: ID Pain. Curr. Med. Res. Opin. 2006, 22, 1555–1565. [Google Scholar] [CrossRef]
  19. Gilron, I.; Baron, R.; Jensen, T. Neuropathic pain: Principles of diagnosis and treatment. Mayo Clin. Proc. 2015, 90, 532–545. [Google Scholar] [CrossRef] [Green Version]
  20. Bernetti, A.; Agostini, F.; de Sire, A.; Mangone, M.; Tognolo, L.; Di Cesare, A.; Ruiu, P.; Paolucci, T.; Invernizzi, M.; Paoloni, M. Neuropathic Pain and Rehabilitation: A Systematic Review of International Guidelines. Diagnostics 2021, 11, 74. [Google Scholar] [CrossRef]
  21. 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] [Green Version]
  22. Hatch, M.N.; Cushing, T.R.; Carlson, G.D.; Chang, E.Y. Neuropathic pain and SCI: Identification and treatment strategies in the 21st century. J. Neurol. Sci. 2018, 384, 75–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Galer, B.S.; Jensen, M.P. Development and preliminary validation of a pain measure specific to neuropathic pain: The Neuropathic Pain Scale. Neurology 1997, 48, 332–338. [Google Scholar] [CrossRef] [PubMed]
  24. Bouhassira, D.; Attal, N.; Fermanian, J.; Alchaar, H.; Gautron, M.; Masquelier, E.; Rostaing, S.; Lanteri-Minet, M.; Collin, E.; Grisart, J.; et al. Development and validation of the Neuropathic Pain Symptom Inventory. Pain 2004, 108, 248–257. [Google Scholar] [CrossRef] [PubMed]
  25. Dworkin, R.H.; Turk, D.C.; Revicki, D.A.; Harding, G.; Coyne, K.S.; Peirce-Sandner, S.; Bhagwat, D.; Everton, D.; Burke, L.B.; Cowan, P.; et al. Development and initial validation of an expanded and revised version of the Short-form McGill Pain Questionnaire (SF-MPQ-2). Pain 2009, 144, 35–42. [Google Scholar] [CrossRef]
  26. Jensen, M.P.; Gammaitoni, A.R.; Olaleye, D.O.; Oleka, N.; Nalamachu, S.R.; Galer, B.S. The pain quality assessment scale: Assessment of pain quality in carpal tunnel syndrome. J. Pain 2006, 7, 823–832. [Google Scholar] [CrossRef]
  27. Martínez-Navarro, M.; Maldonado, R.; Baños, J.E. Why mu-opioid agonists have less analgesic efficacy in neuropathic pain? Eur. J. Pain 2019, 23, 435–454. [Google Scholar] [CrossRef]
  28. Santoni, A.; Mercadante, S.; Arcuri, E. Chronic cancer and non-cancer pain and opioid-induced hyperalgesia share common mechanisms: Neuroinflammation and central sensitization. Minerva Anestesiol. 2021, 87, 210–222. [Google Scholar] [CrossRef]
  29. Cervero, F.; Laird, J.M.A. Mechanisms of touch-evoked pain (allodynia): A new model. Pain 1996, 68, 13–23. [Google Scholar] [CrossRef]
  30. Gao, Y.J.; Zhang, L.; Samad, O.A.; Suter, M.R.; Yasuhiko, K.; Xu, Z.Z.; Park, J.Y.; Lind, A.L.; Ma, Q.; Ji, R.R. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J. Neurosci. 2009, 29, 4096–4108. [Google Scholar] [CrossRef] [Green Version]
  31. Ji, R.R.; Xu, Z.Z.; Gao, Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014, 13, 533–548. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, Z.J.; Cao, D.L.; Zhang, X.; Ji, R.R.; Gao, Y.J. Chemokine contribution to neuropathic pain: Respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. Pain 2013, 154, 2185–2197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Grace, P.M.; Hutchinson, M.R.; Maier, S.F.; Watkins, L.R. Pathological pain and the neuroimmune interface. Nat. Rev. Immunol. 2014, 14, 217–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, Z.J.; Jiang, B.C.; Gao, Y.J. Chemokines in neuron-glial cell interaction and pathogenesis of neuropathic pain. Cell. Mol. Life Sci. 2017, 74, 3275–3291. [Google Scholar] [CrossRef] [PubMed]
  35. Kwiatkowski, K.; Mika, J. The importance of chemokines in neuropathic pain development and opioid analgesic potency. Pharmacol. Rep. 2018, 70, 821–830. [Google Scholar] [CrossRef]
  36. Mélik Parsadaniantz, S.; Rivat, C.; Rostène, W.; Réaux-Le Goazigo, A. Opioid and chemokine receptor crosstalk: A promising target for pain therapy? Nat. Rev. Neurosci. 2015, 16, 69–78. [Google Scholar] [CrossRef]
  37. Steele, A.D.; Szabo, I.; Bednar, F.; Rogers, T.J. Interactions between opioid and chemokine receptors: Heterologous desensitization. Cytokine Growth Factor Rev. 2002, 13, 209–222. [Google Scholar] [CrossRef]
  38. Rogers, T.J. Bidirectional Regulation of Opioid and Chemokine Function. Front. Immunol. 2020, 11, 94. [Google Scholar] [CrossRef] [Green Version]
  39. Woolf, C.J.; Mannion, R.J. Neuropathic pain: Aetiology, symptoms, mechanisms, and management. Lancet 1999, 353, 1959–1964. [Google Scholar] [CrossRef]
  40. Colleoni, M.; Sacerdote, P. Murine models of human neuropathic pain. Biochim. Biophys. Acta 2010, 1802, 924–933. [Google Scholar] [CrossRef] [Green Version]
  41. Jaggi, A.S.; Jain, V.; Singh, N. Animal models of neuropathic pain. Fundam. Clin. Pharmacol. 2011, 25, 1–28. [Google Scholar] [CrossRef]
  42. Wasserman, J.K.; Koeberle, P.D. Development and characterization of a hemorrhagic rat model of central post-stroke pain. Neuroscience 2009, 161, 173–183. [Google Scholar] [CrossRef] [PubMed]
  43. Siddall, P.; Xu, C.L.; Cousins, M. Allodynia following traumatic spinal cord injury in the rat. Neuroreport 1995, 6, 1241–1244. [Google Scholar] [CrossRef] [PubMed]
  44. Aanonsen, L.M.; Wilcox, G.L. Muscimol, gamma-aminobutyric acidA receptors and excitatory amino acids in the mouse spinal cord. J. Pharmacol. Exp. Ther. 1989, 248, 1034–1038. [Google Scholar] [PubMed]
  45. Yezierski, R.P.; Park, S.H. The mechanosensitivity of spinal sensory neurons following intraspinal injections of quisqualic acid in the rat. Neurosci. Lett. 1993, 157, 115–119. [Google Scholar] [CrossRef]
  46. Gaviria, M.; Haton, H.; Sandillon, F.; Privat, A. A mouse model of acute ischemic spinal cord injury. J. Neurotrauma 2002, 19, 205–221. [Google Scholar] [CrossRef]
  47. Watson, B.D.; Prado, R.; Dalton Dietrich, W.; Ginsberg, M.D.; Green, B.A. Photochemically induced spinal cord injury in the rat. Brain Res. 1986, 367, 296–300. [Google Scholar] [CrossRef]
  48. Christensen, M.D.; Everhart, A.W.; Pickelman, J.T.; Hulsebosch, C.E. Mechanical and thermal allodynia in chronic central pain following spinal cord injury. Pain 1996, 68, 97–107. [Google Scholar] [CrossRef]
  49. Wall, P.D.; Devor, M.; Inbal, R.; Scadding, J.W.; Schonfeld, D.; Seltzer, Z.; Tomkiewicz, M.M. Autotomy following peripheral nerve lesions: Experimental anaesthesia dolorosa. Pain 1979, 7, 103–113. [Google Scholar] [CrossRef]
  50. Quintão, N.L.; Balz, D.; Santos, A.R.; Campos, M.M.; Calixto, J.B. Long-lasting neuropathic pain induced by brachial plexus injury in mice: Role triggered by the pro-inflammatory cytokine, tumour necrosis factor alpha. Neuropharmacology 2006, 50, 614–620. [Google Scholar] [CrossRef]
  51. Rodrigues-Filho, R.; Santos, A.R.; Bertelli, J.A.; Calixto, J.B. Avulsion injury of the rat brachial plexus triggers hyperalgesia and allodynia in the hindpaws: A new model for the study of neuropathic pain. Brain Res. 2003, 982, 186–194. [Google Scholar] [CrossRef]
  52. Na, H.S.; Han, J.S.; Ko, K.H.; Hong, S.K. A behavioral model for peripheral neuropathy produced in rat’s tail by inferior caudal trunk injury. Neurosci. Lett. 1994, 177, 50–52. [Google Scholar] [CrossRef]
  53. Lee, B.H.; Won, R.; Baik, E.J.; Lee, S.H.; Moon, C.H. An animal model of neuropathic pain employing injury to the sciatic nerve branches. Neuroreport 2000, 11, 657–661. [Google Scholar] [CrossRef] [PubMed]
  54. Bennett, G.J.; Xie, Y.K. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33, 87–107. [Google Scholar] [CrossRef]
  55. Seltzer, Z.; Dubner, R.; Shir, Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990, 43, 205–218. [Google Scholar] [CrossRef]
  56. Ho Kim, S.; Mo Chung, J. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 1992, 50, 355–363. [Google Scholar] [CrossRef]
  57. Kiso, T.; Watabiki, T.; Tsukamoto, M.; Okabe, M.; Kagami, M.; Nishimura, K.; Aoki, T.; Matsuoka, N. Pharmacological characterization and gene expression profiling of an L5/L6 spinal nerve ligation model for neuropathic pain in mice. Neuroscience 2008, 153, 492–500. [Google Scholar] [CrossRef]
  58. Decosterd, I.; Woolf, C.J. Spared nerve injury: An animal model of persistent peripheral neuropathic pain. Pain 2000, 87, 149–158. [Google Scholar] [CrossRef]
  59. Shields, S.D.; Eckert, W.A., 3rd; Basbaum, A.I. Spared nerve injury model of neuropathic pain in the mouse: A behavioral and anatomic analysis. J. Pain 2003, 4, 465–470. [Google Scholar] [CrossRef]
  60. Walczak, J.S.; Pichette, V.; Leblond, F.; Desbiens, K.; Beaulieu, P. Behavioral, pharmacological and molecular characterization of the saphenous nerve partial ligation: A new model of neuropathic pain. Neuroscience 2005, 132, 1093–1102. [Google Scholar] [CrossRef]
  61. Hulse, R.; Wynick, D.; Donaldson, L.F. Characterization of a novel neuropathic pain model in mice. Neuroreport 2008, 19, 825–829. [Google Scholar] [CrossRef]
  62. Chacur, M.; Milligan, E.D.; Gazda, L.S.; Armstrong, C.; Wang, H.; Tracey, K.J.; Maier, S.F.; Watkins, L.R. A new model of sciatic inflammatory neuritis (SIN): Induction of unilateral and bilateral mechanical allodynia following acute unilateral peri-sciatic immune activation in rats. Pain 2001, 94, 231–244. [Google Scholar] [CrossRef]
  63. Gazda, L.S.; Milligan, E.D.; Hansen, M.K.; Twining, C.M.; Poulos, N.M.; Chacur, M.; O’Connor, K.A.; Armstrong, C.; Maier, S.F.; Watkins, L.R.; et al. Sciatic inflammatory neuritis (SIN): Behavioral allodynia is paralleled by peri-sciatic proinflammatory cytokine and superoxide production. J. Peripher. Nerv. Syst. 2001, 6, 111–129. [Google Scholar] [CrossRef] [PubMed]
  64. Wagner, R.; Myers, R.R. Endoneurial injection of TNF-alpha produces neuropathic pain behaviors. Neuroreport 1996, 7, 2897–2901. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef] [PubMed]
  66. Benbouzid, M.; Pallage, V.; Rajalu, M.; Waltisperger, E.; Doridot, S.; Poisbeau, P.; Freund-Mercier, M.J.; Barrot, M. Sciatic nerve cuffing in mice: A model of sustained neuropathic pain. Eur. J. Pain 2008, 12, 591–599. [Google Scholar] [CrossRef]
  67. Mosconi, T.; Kruger, L. Fixed-diameter polyethylene cuffs applied to the rat sciatic nerve induce a painful neuropathy: Ultrastructural morphometric analysis of axonal alterations. Pain 1996, 64, 37–57. [Google Scholar] [CrossRef]
  68. DeLeo, J.A.; Coombs, D.W.; Willenbring, S.; Colburn, R.W.; Fromm, C.; Wagner, R.; Twitchcll, B.B. Characterization of a neuropathic pain model: Sciatic cryoneurolysis in the rat. Pain 1994, 56, 9–16. [Google Scholar] [CrossRef]
  69. Chiang, H.Y.; Chen, C.T.; Chien, H.F.; Hsieh, S.T. Skin denervation, neuropathology, and neuropathic pain in a laser-induced focal neuropathy. Neurobiol. Dis. 2005, 18, 40–53. [Google Scholar] [CrossRef]
  70. Vadakkan, K.I.; Jia, Y.H.; Zhuo, M. A behavioral model of neuropathic pain induced by ligation of the common peroneal nerve in mice. J. Pain 2005, 6, 747–756. [Google Scholar] [CrossRef]
  71. Imamura, Y.; Kawamoto, H.; Nakanishi, O. Characterization of heat-hyperalgesia in an experimental trigeminal neuropathy in rats. Exp. Brain Res. 1997, 116, 97–103. [Google Scholar] [CrossRef]
  72. Xu, M.; Aita, M.; Chavkin, C. Partial infraorbital nerve ligation as a model of trigeminal nerve injury in the mouse: Behavioral, neural, and glial reactions. J. Pain 2008, 9, 1036–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Roveroni, R.C.; Parada, C.A.; Cecília, M.; Veiga, F.A.; Tambeli, C.H. Development of a behavioral model of TMJ pain in rats: The TMJ formalin test. Pain 2001, 94, 185–191. [Google Scholar] [CrossRef]
  74. Vahidy, W.H.; Ong, W.Y.; Farooqui, A.A.; Yeo, J.F. Effects of intracerebroventricular injections of free fatty acids, lysophospholipids, or platelet activating factor in a mouse model of orofacial pain. Exp. Brain Res. 2006, 174, 781–785. [Google Scholar] [CrossRef] [PubMed]
  75. Aicher, S.A.; Silverman, M.B.; Winkler, C.W.; Bebo, B.F., Jr. Hyperalgesia in an animal model of multiple sclerosis. Pain 2004, 110, 560–570. [Google Scholar] [CrossRef]
  76. Lynch, J.L.; Gallus, N.J.; Ericson, M.E.; Beitz, A.J. Analysis of nociception, sex and peripheral nerve innervation in the TMEV animal model of multiple sclerosis. Pain 2008, 136, 293–304. [Google Scholar] [CrossRef] [Green Version]
  77. Fleetwood-Walker, S.M.; Quinn, J.P.; Wallace, C.; Blackburn-Munro, G.; Kelly, B.G.; Fiskerstrand, C.E.; Nash, A.A.; Dalziel, R.G. Behavioural changes in the rat following infection with varicella-zoster virus. J. Gen. Virol. 1999, 80, 2433–2436. [Google Scholar] [CrossRef] [Green Version]
  78. Takasaki, I.; Andoh, T.; Nitta, M.; Takahata, H.; Nemoto, H.; Shiraki, K.; Nojima, H.; Kuraishi, Y. Pharmacological and immunohistochemical characterization of a mouse model of acute herpetic pain. Jpn. J. Pharmacol. 2000, 83, 319–326. [Google Scholar] [CrossRef]
  79. Takasaki, I.; Andoh, T.; Shiraki, K.; Kuraishi, Y. Allodynia and hyperalgesia induced by herpes simplex virus type-1 infection in mice. Pain 2000, 86, 95–101. [Google Scholar] [CrossRef]
  80. Pan, H.L.; Khan, G.M.; Alloway, K.D.; Chen, S.R. Resiniferatoxin induces paradoxical changes in thermal and mechanical sensitivities in rats: Mechanism of action. J. Neurosci. 2003, 23, 2911–2919. [Google Scholar] [CrossRef] [Green Version]
  81. Herzberg, U.; Sagen, J. Peripheral nerve exposure to HIV viral envelope protein gp120 induces neuropathic pain and spinal gliosis. J. Neuroimmunol. 2001, 116, 29–39. [Google Scholar] [CrossRef]
  82. Keswani, S.C.; Jack, C.; Zhou, C.; Höke, A. Establishment of a rodent model of HIV-associated sensory neuropathy. J. Neurosci. 2006, 26, 10299–10304. [Google Scholar] [CrossRef] [PubMed]
  83. Asai, H.; Ozaki, N.; Shinoda, M.; Nagamine, K.; Tohnai, I.; Ueda, M.; Sugiura, Y. Heat and mechanical hyperalgesia in mice model of cancer pain. Pain 2005, 117, 19–29. [Google Scholar] [CrossRef] [PubMed]
  84. Medhurst, S.J.; Walker, K.; Bowes, M.; Kidd, B.L.; Glatt, M.; Muller, M.; Hattenberger, M.; Vaxelaire, J.; O’Reilly, T.; Wotherspoon, G.; et al. A rat model of bone cancer pain. Pain 2002, 96, 129–140. [Google Scholar] [CrossRef]
  85. Shimoyama, M.; Tanaka, K.; Hasue, F.; Shimoyama, N. A mouse model of neuropathic cancer pain. Pain 2002, 99, 167–174. [Google Scholar] [CrossRef]
  86. Schwei, M.J.; Honore, P.; Rogers, S.D.; Salak-Johnson, J.L.; Finke, M.P.; Ramnaraine, M.L.; Clohisy, D.R.; Mantyh, P.W. Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J. Neurosci. 1999, 19, 10886–10897. [Google Scholar] [CrossRef] [Green Version]
  87. Courteix, C.; Eschalier, A.; Lavarenne, J. Streptozocin-induced diabetic rats: Behavioural evidence for a model of chronic pain. Pain 1993, 53, 81–88. [Google Scholar] [CrossRef]
  88. Morita, K.; Motoyama, N.; Kitayama, T.; Morioka, N.; Kifune, K.; Dohi, T. Spinal antiallodynia action of glycine transporter inhibitors in neuropathic pain models in mice. J. Pharmacol. Exp. Ther. 2008, 326, 633–645. [Google Scholar] [CrossRef] [Green Version]
  89. Lenzen, S. The mechanisms of alloxan- and streptozotocin-induced diabetes. Diabetologia 2008, 51, 216–226. [Google Scholar] [CrossRef] [Green Version]
  90. Goto, Y.; Suzuki, K.; Ono, T.; Sasaki, M.; Toyota, T. Development of diabetes in the non-obese NIDDM rat (GK rat). Adv. Exp. Med. Biol. 1988, 246, 29–31. [Google Scholar] [CrossRef]
  91. Norido, F.; Canella, R.; Zanoni, R.; Gorio, A. Development of diabetic neuropathy in the C57BL/Ks (db/db) mouse and its treatment with gangliosides. Exp. Neurol. 1984, 83, 221–232. [Google Scholar] [CrossRef]
  92. Mosseri, R.; Waner, T.; Shefi, M.; Shafrir, E.; Meyerovitch, J. Gluconeogenesis in non-obese diabetic (NOD) mice: In vivo effects of vandadate treatment on hepatic glucose-6-phoshatase and phosphoenolpyruvate carboxykinase. Metabolism 2000, 49, 321–325. [Google Scholar] [CrossRef]
  93. Meyerovitch, J.; Rothenberg, P.; Shechter, Y.; Bonner-Weir, S.; Kahn, C.R. Vanadate normalizes hyperglycemia in two mouse models of non-insulin-dependent diabetes mellitus. J. Clin. Investig. 1991, 87, 1286–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Authier, N.; Gillet, J.P.; Fialip, J.; Eschalier, A.; Coudore, F. A new animal model of vincristine-induced nociceptive peripheral neuropathy. Neurotoxicology 2003, 24, 797–805. [Google Scholar] [CrossRef]
  95. Szilvássy, J.; Sziklai, I.; Racz, T.; Horvath, P.; Rabloczky, G.; Szilvassy, Z. Impaired bronchomotor responses to field stimulation in guinea-pigs with cisplatin-induced neuropathy. Eur. J. Pharmacol. 2000, 403, 259–265. [Google Scholar] [CrossRef]
  96. Cavaletti, G.; Tredici, G.; Petruccioli, M.G.; Dondè, E.; Tredici, P.; Marmiroli, P.; Minoia, C.; Ronchi, A.; Bayssas, M.; Etienne, G.G. Effects of different schedules of oxaliplatin treatment on the peripheral nervous system of the rat. Eur. J. Cancer 2001, 37, 2457–2463. [Google Scholar] [CrossRef]
  97. Flatters, S.J.L.; Bennett, G.J. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: Evidence for mitochondrial dysfunction. Pain 2006, 122, 245–257. [Google Scholar] [CrossRef] [Green Version]
  98. Anderson, T.D.; Davidovich, A.; Arceo, R.; Brosnan, C.; Arezzo, J.; Schaumburg, H. Peripheral neuropathy induced by 2′,3′-dideoxycytidine. A rabbit model of 2′,3′-dideoxycytidine neurotoxicity. Lab. Investig. 1992, 66, 63–74. [Google Scholar]
  99. Schmued, L.C.; Albertson, C.M.; Andrews, A.; Sandberg, J.A.; Nickols, J.; Slikker, W., Jr. Evaluation of brain and nerve pathology in rats chronically dosed with ddI or isoniazid. Neurotoxicol. Teratol. 1996, 18, 555–563. [Google Scholar] [CrossRef]
  100. Suter, U.; Moskow, J.J.; Welcher, A.A.; Snipes, G.J.; Kosaras, B.; Sidman, R.L.; Buchberg, A.M.; Shooter, E.M. A leucine-to-proline mutation in the putative first transmembrane domain of the 22-kDa peripheral myelin protein in the trembler-J mouse. Proc. Natl. Acad. Sci. USA 1992, 89, 4382–4386. [Google Scholar] [CrossRef] [Green Version]
  101. Adlkofer, K.; Martini, R.; Aguzzi, A.; Zielasek, J.; Toyka, K.V.; Suter, U. Hypermyelination and demyelinating peripheral neuropathy in Pmp22-deficient mice. Nat. Genet. 1995, 11, 274–280. [Google Scholar] [CrossRef]
  102. Adlkofer, K.; Frei, R.; Neuberg, D.H.; Zielasek, J.; Toyka, K.V.; Suter, U. Heterozygous peripheral myelin protein 22-deficient mice are affected by a progressive demyelinating tomaculous neuropathy. J. Neurosci. 1997, 17, 4662–4671. [Google Scholar] [CrossRef] [PubMed]
  103. Martini, R. Animal models for inherited peripheral neuropathies. J. Anat. 1997, 191, 321–336. [Google Scholar] [CrossRef] [PubMed]
  104. Ahuja, C.S.; Wilson, J.R.; Nori, S.; Kotter, M.R.N.; Druschel, C.; Curt, A.; Fehlings, M.G. Traumatic spinal cord injury. Nat. Rev. Dis. Primers 2017, 3, 17018. [Google Scholar] [CrossRef] [PubMed]
  105. Kuner, R.; Flor, H. Structural plasticity and reorganisation in chronic pain. Nat. Rev. Neurosci. 2016, 18, 20–30. [Google Scholar] [CrossRef]
  106. Latremoliere, A.; Woolf, C.J. Central sensitization: A generator of pain hypersensitivity by central neural plasticity. J. Pain 2009, 10, 895–926. [Google Scholar] [CrossRef] [Green Version]
  107. Kuner, R. Central mechanisms of pathological pain. Nat. Med. 2010, 16, 1258–1266. [Google Scholar] [CrossRef]
  108. Kang, S.J.; Kwak, C.; Lee, J.; Sim, S.E.; Shim, J.; Choi, T.; Collingridge, G.L.; Zhuo, M.; Kaang, B.K. Bidirectional modulation of hyperalgesia via the specific control of excitatory and inhibitory neuronal activity in the ACC. Mol. Brain 2015, 8, 81. [Google Scholar] [CrossRef] [Green Version]
  109. Liu, M.; Wood, J.N. The roles of sodium channels in nociception: Implications for mechanisms of neuropathic pain. Pain Med. 2011, 12, S93–S99. [Google Scholar] [CrossRef] [Green Version]
  110. Todorovic, S.M.; Jevtovic-Todorovic, V. T-type voltage-gated calcium channels as targets for the development of novel pain therapies. Br. J. Pharmacol. 2011, 163, 484–495. [Google Scholar] [CrossRef] [Green Version]
  111. Tsantoulas, C. Emerging potassium channel targets for the treatment of pain. Curr. Opin. Support. Palliat. Care 2015, 9, 147–154. [Google Scholar] [CrossRef]
  112. Baron, R.; Binder, A.; Wasner, G. Neuropathic pain: Diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol. 2010, 9, 807–819. [Google Scholar] [CrossRef]
  113. Nickel, F.T.; Seifert, F.; Lanz, S.; Maihöfner, C. Mechanisms of neuropathic pain. Eur. Neuropsychopharmacol. 2012, 22, 81–91. [Google Scholar] [CrossRef] [PubMed]
  114. Ji, R.R.; Berta, T.; Nedergaard, M. Glia and pain: Is chronic pain a gliopathy? Pain 2013, 154 (Suppl. S1), S10–S28. [Google Scholar] [CrossRef] [PubMed]
  115. Malcangio, M. Role of the immune system in neuropathic pain. Scand. J. Pain 2019, 20, 33–37. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, G.; Luo, X.; Qadri, M.Y.; Berta, T.; Ji, R.R. Sex-Dependent Glial Signaling in Pathological Pain: Distinct Roles of Spinal Microglia and Astrocytes. Neurosci. Bull. 2018, 34, 98–108. [Google Scholar] [CrossRef] [Green Version]
  117. Wei, Z.; Fei, Y.; Su, W.; Chen, G. Emerging Role of Schwann Cells in Neuropathic Pain: Receptors, Glial Mediators and Myelination. Front. Cell Neurosci. 2019, 13, 116. [Google Scholar] [CrossRef] [Green Version]
  118. Ji, R.R.; Chamessian, A.; Zhang, Y.Q. Pain regulation by non-neuronal cells and inflammation. Science 2016, 354, 572–577. [Google Scholar] [CrossRef] [Green Version]
  119. Kiguchi, N.; Kobayashi, Y.; Kishioka, S. Chemokines and cytokines in neuroinflammation leading to neuropathic pain. Curr. Opin. Pharmacol. 2012, 12, 55–61. [Google Scholar] [CrossRef]
  120. Ji, R.R.; Nackley, A.; Huh, Y.; Terrando, N.; Maixner, W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018, 129, 343–366. [Google Scholar] [CrossRef]
  121. 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] [Green Version]
  122. Bajetto, A.; Bonavia, R.; Barbero, S.; Schettini, G. Characterization of chemokines and their receptors in the central nervous system: Physiopathological implications. J. Neurochem. 2002, 82, 1311–1329. [Google Scholar] [CrossRef] [PubMed]
  123. Williams, J.L.; Holman, D.W.; Klein, R.S. Chemokines in the balance: Maintenance of homeostasis and protection at CNS barriers. Front. Cell Neurosci. 2014, 8, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. 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]
  125. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef] [Green Version]
  126. Cartier, L.; Hartley, O.; Dubois-Dauphin, M.; Krause, K.H. Chemokine receptors in the central nervous system: Role in brain inflammation and neurodegenerative diseases. Brain. Res. Brain. Res. Rev. 2005, 48, 16–42. [Google Scholar] [CrossRef]
  127. Miller, R.J.; Rostene, W.; Apartis, E.; Banisadr, G.; Biber, K.; Milligan, E.D.; White, F.A.; Zhang, J. Chemokine action in the nervous system. J. Neurosci. 2008, 28, 11792–11795. [Google Scholar] [CrossRef]
  128. Moser, B.; Wolf, M.; Walz, A.; Loetscher, P. Chemokines: Multiple levels of leukocyte migration control. Trends Immunol. 2004, 25, 75–84. [Google Scholar] [CrossRef]
  129. Asensio, V.C.; Campbell, I.L. Chemokines in the CNS: Plurifunctional mediators in diverse states. Trends Neurosci. 1999, 22, 504–512. [Google Scholar] [CrossRef]
  130. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef]
  131. Ubogu, E.E.; Cossoy, M.B.; Ransohoff, R.M. The expression and function of chemokines involved in CNS inflammation. Trends Pharmacol. Sci. 2006, 27, 48–55. [Google Scholar] [CrossRef]
  132. Savarin-Vuaillat, C.; Ransohoff, R.M. Chemokines and chemokine receptors in neurological disease: Raise, retain, or reduce? Neurotherapeutics 2007, 4, 590–601. [Google Scholar] [CrossRef]
  133. Romagnani, P.; Lasagni, L.; Annunziato, F.; Serio, M.; Romagnani, S. CXC chemokines: The regulatory link between inflammation and angiogenesis. Trends Immunol. 2004, 25, 201–209. [Google Scholar] [CrossRef] [PubMed]
  134. 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] [PubMed]
  135. Proudfoot, A.E. Chemokine receptors: Multifaceted therapeutic targets. Nat. Rev. Immunol. 2002, 2, 106–115. [Google Scholar] [CrossRef]
  136. Montague, K.; Malcangio, M. The therapeutic potential of targeting chemokine signalling in the treatment of chronic pain. J. Neurochem. 2017, 141, 520–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Van Steenwinckel, J.; Reaux-Le Goazigo, A.; Pommier, B.; Mauborgne, A.; Dansereau, M.A.; Kitabgi, P.; Sarret, P.; Pohl, M.; Mélik Parsadaniantz, S. CCL2 released from neuronal synaptic vesicles in the spinal cord is a major mediator of local inflammation and pain after peripheral nerve injury. J. Neurosci. 2011, 31, 5865–5875. [Google Scholar] [CrossRef]
  138. Zhu, X.; Cao, S.; Zhu, M.D.; Liu, J.Q.; Chen, J.J.; Gao, Y.J. Contribution of chemokine CCL2/CCR2 signaling in the dorsal root ganglion and spinal cord to the maintenance of neuropathic pain in a rat model of lumbar disc herniation. J. Pain 2014, 15, 516–526. [Google Scholar] [CrossRef] [PubMed]
  139. Illias, A.M.; Gist, A.C.; Zhang, H.; Kosturakis, A.K.; Dougherty, P.M. Chemokine CCL2 and its receptor CCR2 in the dorsal root ganglion contribute to oxaliplatin-induced mechanical hypersensitivity. Pain 2018, 159, 1308–1316. [Google Scholar] [CrossRef]
  140. Zhang, J.; Shi, X.Q.; Echeverry, S.; Mogil, J.S.; De Koninck, Y.; Rivest, S. Expression of CCR2 in both resident and bone marrow-derived microglia plays a critical role in neuropathic pain. J. Neurosci. 2007, 27, 12396–12406. [Google Scholar] [CrossRef]
  141. Zhang, J.; De Koninck, Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J. Neurochem. 2006, 97, 772–783. [Google Scholar] [CrossRef]
  142. Dubový, P.; Klusáková, I.; Hradilová-Svíženská, I.; Joukal, M.; Boadas-Vaello, P. Activation of Astrocytes and Microglial Cells and CCL2/CCR2 Upregulation in the Dorsolateral and Ventrolateral Nuclei of Periaqueductal Gray and Rostral Ventromedial Medulla Following Different Types of Sciatic Nerve Injury. Front. Cell Neurosci. 2018, 12, 40. [Google Scholar] [CrossRef]
  143. 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]
  144. Hu, Y.; Kodithuwakku, N.D.; Zhou, L.; Li, C.; Han, D.; Fang, W.; Liu, J.; Li, Y. Levo-Corydalmine Alleviates Neuropathic Cancer Pain Induced by Tumor Compression via the CCL2/CCR2 Pathway. Molecules 2017, 22, 937. [Google Scholar] [CrossRef] [Green Version]
  145. Lindia, J.A.; McGowan, E.; Jochnowitz, N.; Abbadie, C. Induction of CX3CL1 expression in astrocytes and CX3CR1 in microglia in the spinal cord of a rat model of neuropathic pain. J. Pain 2005, 6, 434–438. [Google Scholar] [CrossRef]
  146. Zhuang, Z.Y.; Kawasaki, Y.; Tan, P.H.; Wen, Y.R.; Huang, J.; Ji, R.R. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain Behav. Immun. 2007, 21, 642–651. [Google Scholar] [CrossRef] [Green Version]
  147. Milligan, E.D.; Zapata, V.; Chacur, M.; Schoeniger, D.; Biedenkapp, J.; O’Connor, K.A.; Verge, G.M.; Chapman, G.; Green, P.; Foster, A.C.; et al. Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats. Eur. J. Neurosci. 2004, 20, 2294–2302. [Google Scholar] [CrossRef]
  148. Staniland, A.A.; Clark, A.K.; Wodarski, R.; Sasso, O.; Maione, F.; D’Acquisto, F.; Malcangio, M. Reduced inflammatory and neuropathic pain and decreased spinal microglial response in fractalkine receptor (CX3CR1) knockout mice. J. Neurochem. 2010, 114, 1143–1157. [Google Scholar] [CrossRef]
  149. Bäckryd, E.; Lind, A.L.; Thulin, M.; Larsson, A.; Gerdle, B.; Gordh, T. High levels of cerebrospinal fluid chemokines point to the presence of neuroinflammation in peripheral neuropathic pain: A cross-sectional study of 2 cohorts of patients compared with healthy controls. Pain 2017, 158, 2487–2495. [Google Scholar] [CrossRef]
  150. Teodorczyk-Injeyan, J.A.; McGregor, M.; Triano, J.J.; Injeyan, S.H. Elevated Production of Nociceptive CC Chemokines and sE-Selectin in Patients With Low Back Pain and the Effects of Spinal Manipulation: A Nonrandomized Clinical Trial. Clin. J. Pain 2018, 34, 68–75. [Google Scholar] [CrossRef] [Green Version]
  151. Jönsson, M.; Gerdle, B.; Ghafouri, B.; Bäckryd, E. The inflammatory profile of cerebrospinal fluid, plasma, and saliva from patients with severe neuropathic pain and healthy controls-a pilot study. BMC Neurosci. 2021, 22, 6. [Google Scholar] [CrossRef]
  152. Mordillo-Mateos, L.; Sánchez-Ramos, A.; Coperchini, F.; Bustos-Guadamillas, I.; Alonso-Bonilla, C.; Vargas-Baquero, E.; Rodriguez-Carrión, I.; Rotondi, M.; Oliviero, A. Development of chronic pain in males with traumatic spinal cord injury: Role of circulating levels of the chemokines CCL2 and CXCL10 in subacute stage. Spinal. Cord. 2019, 57, 953–959. [Google Scholar] [CrossRef] [PubMed]
  153. Negri, L.; Lattanzi, R.; Giannini, E.; Canestrelli, M.; Nicotra, A.; Melchiorri, P. Bv8/Prokineticins and their Receptors A New Pronociceptive System. Int. Rev. Neurobiol. 2009, 85, 145–157. [Google Scholar] [CrossRef] [PubMed]
  154. Negri, L.; Lattanzi, R. Bv8/PK2 and prokineticin receptors: A druggable pronociceptive system. Curr. Opin. Pharmacol. 2012, 12, 62–66. [Google Scholar] [CrossRef] [PubMed]
  155. Maftei, D.; Marconi, V.; Florenzano, F.; Giancotti, L.A.; Castelli, M.; Moretti, S.; Borsani, E.; Rodella, L.F.; Balboni, G.; Luongo, L.; et al. Controlling the activation of the Bv8/prokineticin system reduces neuroinflammation and abolishes thermal and tactile hyperalgesia in neuropathic animals. Br. J. Pharmacol. 2014, 171, 4850–4865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Castelli, M.; Amodeo, G.; Negri, L.; Lattanzi, R.; Maftei, D.; Gotti, C.; Pistillo, F.; Onnis, V.; Congu, C.; Panerai, A.E.; et al. Antagonism of the Prokineticin System Prevents and Reverses Allodynia and Inflammation in a Mouse Model of Diabetes. PLoS ONE 2016, 11, e0146259. [Google Scholar] [CrossRef]
  157. Moschetti, G.; Amodeo, G.; Maftei, D.; Lattanzi, R.; Procacci, P.; Sartori, P.; Balboni, G.; Onnis, V.; Conte, V.; Panerai, A.; et al. Targeting prokineticin system counteracts hypersensitivity, neuroinflammation, and tissue damage in a mouse model of bortezomib-induced peripheral neuropathy. J. Neuroinflamm. 2019, 16, 89. [Google Scholar] [CrossRef] [Green Version]
  158. Xie, L.; Yin, Y.; Benowitz, L. Chemokine CCL5 promotes robust optic nerve regeneration and mediates many of the effects of CNTF gene therapy. Proc. Natl. Acad. Sci. USA 2021, 118, e2017282118. [Google Scholar] [CrossRef]
  159. Dedoni, S.; Campbell, L.A.; Harvey, B.K.; Avdoshina, V.; Mocchetti, I. The orphan G-protein-coupled receptor 75 signaling is activated by the chemokine CCL5. J. Neurochem. 2018, 146, 526–539. [Google Scholar] [CrossRef]
  160. Lattanzi, R.; Severini, C.; Maftei, D.; Saso, L.; Badiani, A. The Role of Prokineticin 2 in Oxidative Stress and in Neuropathological Processes. Front. Pharmacol. 2021, 12, 640441. [Google Scholar] [CrossRef]
  161. Fields, H. State-dependent opioid control of pain. Nat. Rev. Neurosci. 2004, 5, 565–575. [Google Scholar] [CrossRef]
  162. Ossipov, M.H.; Dussor, G.O.; Porreca, F. Central modulation of pain. J. Clin. Investig. 2010, 120, 3779–3787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Matthes, H.W.; Maldonado, R.; Simonin, F.; Valverde, O.; Slowe, S.; Kitchen, I.; Befort, K.; Dierich, A.; Le Meur, M.; Dollé, P.; et al. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene. Nature 1996, 383, 819–823. [Google Scholar] [CrossRef] [PubMed]
  164. Caraceni, A.; Hanks, G.; Kaasa, S.; Bennett, M.I.; Brunelli, C.; Cherny, N.; Dale, O.; De Conno, F.; Fallon, M.; Hanna, M.; et al. Use of opioid analgesics in the treatment of cancer pain: Evidence-based recommendations from the EAPC. Lancet Oncol. 2012, 13, e58–e68. [Google Scholar] [CrossRef]
  165. Allen, S.C. Neuropathic pain—The case for opioid therapy. Oncology 2008, 74, 76–82. [Google Scholar] [CrossRef] [PubMed]
  166. Auret, K.; Schug, S.A. Underutilisation of opioids in elderly patients with chronic pain: Approaches to correcting the problem. Drugs Aging 2005, 22, 641–654. [Google Scholar] [CrossRef]
  167. Arnér, S.; Meyerson, B.A. Lack of analgesic effect of opioids on neuropathic and idiopathic forms of pain. Pain 1988, 33, 11–23. [Google Scholar] [CrossRef]
  168. Cooper, T.E.; Chen, J.; Wiffen, P.J.; Derry, S.; Carr, D.B.; Aldington, D.; Cole, P.; Moore, R.A. Morphine for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2017, 5, Cd011669. [Google Scholar] [CrossRef]
  169. Kupers, R.C.; Konings, H.; Adriaensen, H.; Gybels, J.M. Morphine differentially affects the sensory and affective pain ratings in neurogenic and idiopathic forms of pain. Pain 1991, 47, 5–12. [Google Scholar] [CrossRef]
  170. Rowbotham, M.C.; Reisner-Keller, L.A.; Fields, H.L. Both intravenous lidocaine and morphine reduce the pain of postherpetic neuralgia. Neurology 1991, 41, 1024–1028. [Google Scholar] [CrossRef]
  171. Gaskell, H.; Derry, S.; Stannard, C.; Moore, R.A. Oxycodone for neuropathic pain in adults. Cochrane Database Syst. Rev. 2016, 7, Cd010692. [Google Scholar] [CrossRef]
  172. Gimbel, J.S.; Richards, P.; Portenoy, R.K. Controlled-release oxycodone for pain in diabetic neuropathy: A randomized controlled trial. Neurology 2003, 60, 927–934. [Google Scholar] [CrossRef] [PubMed]
  173. Bleeker, C.P.; Bremer, R.C.; Dongelmans, D.A.; van Dongen, R.T.; Crul, B.J. Inefficacy of high-dose transdermal fentanyl in a patient with neuropathic pain, a case report. Eur. J. Pain 2001, 5, 325–329. [Google Scholar] [CrossRef] [PubMed]
  174. Agarwal, S.; Polydefkis, M.; Block, B.; Haythornthwaite, J.; Raja, S.N. Transdermal fentanyl reduces pain and improves functional activity in neuropathic pain states. Pain Med. 2007, 8, 554–562. [Google Scholar] [CrossRef] [PubMed]
  175. Harati, Y.; Gooch, C.; Swenson, M.; Edelman, S.; Greene, D.; Raskin, P.; Donofrio, P.; Cornblath, D.; Sachdeo, R.; Siu, C.O.; et al. Double-blind randomized trial of tramadol for the treatment of the pain of diabetic neuropathy. Neurology 1998, 50, 1842–1846. [Google Scholar] [CrossRef]
  176. Harati, Y.; Gooch, C.; Swenson, M.; Edelman, S.V.; Greene, D.; Raskin, P.; Donofrio, P.; Cornblath, D.; Olson, W.H.; Kamin, M. Maintenance of the long-term effectiveness of tramadol in treatment of the pain of diabetic neuropathy. J. Diabetes Complicat. 2000, 14, 65–70. [Google Scholar] [CrossRef]
  177. Hollingshead, J.; Dühmke, R.M.; Cornblath, D.R. Tramadol for neuropathic pain. Cochrane Database Syst. Rev. 2006, Cd003726. [Google Scholar] [CrossRef]
  178. Schwartz, S.; Etropolski, M.; Shapiro, D.Y.; Okamoto, A.; Lange, R.; Haeussler, J.; Rauschkolb, C. Safety and efficacy of tapentadol ER in patients with painful diabetic peripheral neuropathy: Results of a randomized-withdrawal, placebo-controlled trial. Curr. Med. Res. Opin. 2011, 27, 151–162. [Google Scholar] [CrossRef]
  179. Apaydin, S.; Uyar, M.; Karabay, N.U.; Erhan, E.; Yegul, I.; Tuglular, I. The antinociceptive effect of tramadol on a model of neuropathic pain in rats. Life Sci. 2000, 66, 1627–1637. [Google Scholar] [CrossRef]
  180. Tzschentke, T.M.; Christoph, T.; Kögel, B.; Schiene, K.; Hennies, H.H.; Englberger, W.; Haurand, M.; Jahnel, U.; Cremers, T.I.; Friderichs, E.; et al. (-)-(1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)-phenol hydrochloride (tapentadol HCl): A novel mu-opioid receptor agonist/norepinephrine reuptake inhibitor with broad-spectrum analgesic properties. J. Pharmacol. Exp. Ther. 2007, 323, 265–276. [Google Scholar] [CrossRef]
  181. Schröder, W.; Vry, J.D.; Tzschentke, T.M.; Jahnel, U.; Christoph, T. Differential contribution of opioid and noradrenergic mechanisms of tapentadol in rat models of nociceptive and neuropathic pain. Eur. J. Pain 2010, 14, 814–821. [Google Scholar] [CrossRef]
  182. Bian, D.; Ossipov, M.H.; Ibrahim, M.; Raffa, R.B.; Tallarida, R.J.; Malan, T.P., Jr.; Lai, J.; Porreca, F. Loss of antiallodynic and antinociceptive spinal/supraspinal morphine synergy in nerve-injured rats: Restoration by MK-801 or dynorphin antiserum. Brain Res. 1999, 831, 55–63. [Google Scholar] [CrossRef]
  183. Lee, Y.W.; Chaplan, S.R.; Yaksh, T.L. Systemic and supraspinal, but not spinal, opiates suppress allodynia in a rat neuropathic pain model. Neurosci. Lett. 1995, 199, 111–114. [Google Scholar] [CrossRef]
  184. Back, S.K.; Lee, J.; Hong, S.K.; Na, H.S. Loss of spinal mu-opioid receptor is associated with mechanical allodynia in a rat model of peripheral neuropathy. Pain 2006, 123, 117–126. [Google Scholar] [CrossRef] [PubMed]
  185. Fundytus, M.E.; Yashpal, K.; Chabot, J.G.; Osborne, M.G.; Lefebvre, C.D.; Dray, A.; Henry, J.L.; Coderre, T.J. Knockdown of spinal metabotropic glutamate receptor 1 (mGluR(1)) alleviates pain and restores opioid efficacy after nerve injury in rats. Br. J. Pharmacol. 2001, 132, 354–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Idänpään-Heikkilä, J.J.; Guilbaud, G.; Kayser, V. Prevention of tolerance to the antinociceptive effects of systemic morphine by a selective cholecystokinin-B receptor antagonist in a rat model of peripheral neuropathy. J. Pharmacol. Exp. Ther. 1997, 282, 1366–1372. [Google Scholar]
  187. Kimura, M.; Obata, H.; Saito, S. Peripheral nerve injury reduces analgesic effects of systemic morphine via spinal 5-hydroxytryptamine 3 receptors. Anesthesiology 2014, 121, 362–371. [Google Scholar] [CrossRef] [Green Version]
  188. Lee, C.Y.; Perez, F.M.; Wang, W.; Guan, X.; Zhao, X.; Fisher, J.L.; Guan, Y.; Sweitzer, S.M.; Raja, S.N.; Tao, Y.X. Dynamic temporal and spatial regulation of mu opioid receptor expression in primary afferent neurons following spinal nerve injury. Eur. J. Pain 2011, 15, 669–675. [Google Scholar] [CrossRef]
  189. Obara, I.; Parkitna, J.R.; Korostynski, M.; Makuch, W.; Kaminska, D.; Przewlocka, B.; Przewlocki, R. Local peripheral opioid effects and expression of opioid genes in the spinal cord and dorsal root ganglia in neuropathic and inflammatory pain. Pain 2009, 141, 283–291. [Google Scholar] [CrossRef]
  190. Ochiai, W.; Kaneta, M.; Nagae, M.; Yuzuhara, A.; Li, X.; Suzuki, H.; Hanagata, M.; Kitaoka, S.; Suto, W.; Kusunoki, Y.; et al. Mice with neuropathic pain exhibit morphine tolerance due to a decrease in the morphine concentration in the brain. Eur. J. Pharm. Sci. 2016, 92, 298–304. [Google Scholar] [CrossRef]
  191. Vanderah, T.W.; Gardell, L.R.; Burgess, S.E.; Ibrahim, M.; Dogrul, A.; Zhong, C.M.; Zhang, E.T.; Malan, T.P., Jr.; Ossipov, M.H.; Lai, J.; et al. Dynorphin promotes abnormal pain and spinal opioid antinociceptive tolerance. J. Neurosci. 2000, 20, 7074–7079. [Google Scholar] [CrossRef] [Green Version]
  192. Ossipov, M.H.; Lopez, Y.; Nichols, M.L.; Bian, D.; Porreca, F. Inhibition by spinal morphine of the tail-flick response is attenuated in rats with nerve ligation injury. Neurosci. Lett. 1995, 199, 83–86. [Google Scholar] [CrossRef]
  193. Rashid, M.H.; Inoue, M.; Toda, K.; Ueda, H. Loss of peripheral morphine analgesia contributes to the reduced effectiveness of systemic morphine in neuropathic pain. J. Pharmacol. Exp. Ther. 2004, 309, 380–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Yamamoto, T.; Sakashita, Y. Differential effects of intrathecally administered morphine and its interaction with cholecystokinin-B antagonist on thermal hyperalgesia following two models of experimental mononeuropathy in the rat. Anesthesiology 1999, 90, 1382–1391. [Google Scholar] [CrossRef] [PubMed]
  195. Zhou, X.L.; Yu, L.N.; Wang, Y.; Tang, L.H.; Peng, Y.N.; Cao, J.L.; Yan, M. Increased methylation of the MOR gene proximal promoter in primary sensory neurons plays a crucial role in the decreased analgesic effect of opioids in neuropathic pain. Mol. Pain 2014, 10, 51. [Google Scholar] [CrossRef] [Green Version]
  196. Grace, P.M.; Strand, K.A.; Galer, E.L.; Maier, S.F.; Watkins, L.R. MicroRNA-124 and microRNA-146a both attenuate persistent neuropathic pain induced by morphine in male rats. Brain Res. 2018, 1692, 9–11. [Google Scholar] [CrossRef]
  197. Grace, P.M.; Strand, K.A.; Galer, E.L.; Rice, K.C.; Maier, S.F.; Watkins, L.R. Protraction of neuropathic pain by morphine is mediated by spinal damage associated molecular patterns (DAMPs) in male rats. Brain. Behav. Immun. 2018, 72, 45–50. [Google Scholar] [CrossRef]
  198. Grace, P.M.; Strand, K.A.; Galer, E.L.; Urban, D.J.; Wang, X.; Baratta, M.V.; Fabisiak, T.J.; Anderson, N.D.; Cheng, K.; Greene, L.I.; et al. Morphine paradoxically prolongs neuropathic pain in rats by amplifying spinal NLRP3 inflammasome activation. Proc. Natl. Acad. Sci. USA 2016, 113, E3441–E3450. [Google Scholar] [CrossRef] [Green Version]
  199. Green-Fulgham, S.M.; Ball, J.B.; Kwilasz, A.J.; Fabisiak, T.; Maier, S.F.; Watkins, L.R.; Grace, P.M. Oxycodone, fentanyl, and morphine amplify established neuropathic pain in male rats. Pain 2019, 160, 2634–2640. [Google Scholar] [CrossRef]
  200. Roeckel, L.A.; Utard, V.; Reiss, D.; Mouheiche, J.; Maurin, H.; Robé, A.; Audouard, E.; Wood, J.N.; Goumon, Y.; Simonin, F.; et al. Morphine-induced hyperalgesia involves mu opioid receptors and the metabolite morphine-3-glucuronide. Sci. Rep. 2017, 7, 10406. [Google Scholar] [CrossRef]
  201. Sánchez-Blázquez, P.; Rodríguez-Muñoz, M.; Berrocoso, E.; Garzón, J. The plasticity of the association between mu-opioid receptor and glutamate ionotropic receptor N in opioid analgesic tolerance and neuropathic pain. Eur. J. Pharmacol. 2013, 716, 94–105. [Google Scholar] [CrossRef] [Green Version]
  202. Minami, K.; Hasegawa, M.; Ito, H.; Nakamura, A.; Tomii, T.; Matsumoto, M.; Orita, S.; Matsushima, S.; Miyoshi, T.; Masuno, K.; et al. Morphine, oxycodone, and fentanyl exhibit different analgesic profiles in mouse pain models. J. Pharmacol. Sci 2009, 111, 60–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. DeLeo, J.A.; Tanga, F.Y.; Tawfik, V.L. Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. Neuroscientist 2004, 10, 40–52. [Google Scholar] [CrossRef] [PubMed]
  204. Hutchinson, M.R.; Coats, B.D.; Lewis, S.S.; Zhang, Y.; Sprunger, D.B.; Rezvani, N.; Baker, E.M.; Jekich, B.M.; Wieseler, J.L.; Somogyi, A.A.; et al. Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav. Immun. 2008, 22, 1178–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. El-Hage, N.; Gurwell, J.A.; Singh, I.N.; Knapp, P.E.; Nath, A.; Hauser, K.F. Synergistic increases in intracellular Ca2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia 2005, 50, 91–106. [Google Scholar] [CrossRef] [Green Version]
  206. Rock, R.B.; Hu, S.; Sheng, W.S.; Peterson, P.K. Morphine stimulates CCL2 production by human neurons. J. Neuroinflamm. 2006, 3, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Wetzel, M.A.; Steele, A.D.; Eisenstein, T.K.; Adler, M.W.; Henderson, E.E.; Rogers, T.J. Mu-opioid induction of monocyte chemoattractant protein-1, RANTES, and IFN-gamma-inducible protein-10 expression in human peripheral blood mononuclear cells. J. Immunol. 2000, 165, 6519–6524. [Google Scholar] [CrossRef]
  208. Liu, L.; Gao, X.J.; Ren, C.G.; Hu, J.H.; Liu, X.W.; Zhang, P.; Zhang, Z.W.; Fu, Z.J. Monocyte chemoattractant protein-1 contributes to morphine tolerance in rats with cancer-induced bone pain. Exp. Ther. Med. 2017, 13, 461–466. [Google Scholar] [CrossRef]
  209. Zhao, C.M.; Guo, R.X.; Hu, F.; Meng, J.L.; Mo, L.Q.; Chen, P.X.; Liao, X.X.; Cui, Y.; Feng, J.Q. Spinal MCP-1 contributes to the development of morphine antinociceptive tolerance in rats. Am. J. Med. Sci 2012, 344, 473–479. [Google Scholar] [CrossRef]
  210. Lin, C.P.; Kang, K.H.; Tu, H.J.; Wu, M.Y.; Lin, T.H.; Liou, H.C.; Sun, W.Z.; Fu, W.M. CXCL12/CXCR4 Signaling Contributes to the Pathogenesis of Opioid Tolerance: A Translational Study. Anesth. Analg. 2017, 124, 972–979. [Google Scholar] [CrossRef]
  211. Lin, C.P.; Lu, D.H. Role of Neuroinflammation in Opioid Tolerance: Translational Evidence from Human-to-Rodent Studies. Adv. Exp. Med. Biol. 2018, 1099, 125–139. [Google Scholar] [CrossRef]
  212. Wang, W.; Peng, Y.; Yang, H.; Bu, H.; Guo, G.; Liu, D.; Shu, B.; Tian, X.; Luo, A.; Zhang, X.; et al. Potential role of CXCL10/CXCR3 signaling in the development of morphine tolerance in periaqueductal gray. Neuropeptides 2017, 65, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lin, C.P.; Kang, K.H.; Lin, T.H.; Wu, M.Y.; Liou, H.C.; Chuang, W.J.; Sun, W.Z.; Fu, W.M. Role of spinal CXCL1 (GROα) in opioid tolerance: A human-to-rodent translational study. Anesthesiology 2015, 122, 666–676. [Google Scholar] [CrossRef] [PubMed]
  214. Chen, X.; Geller, E.B.; Rogers, T.J.; Adler, M.W. The chemokine CX3CL1/fractalkine interferes with the antinociceptive effect induced by opioid agonists in the periaqueductal grey of rats. Brain Res. 2007, 1153, 52–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Johnston, I.N.; Milligan, E.D.; Wieseler-Frank, J.; Frank, M.G.; Zapata, V.; Campisi, J.; Langer, S.; Martin, D.; Green, P.; Fleshner, M.; et al. A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J. Neurosci. 2004, 24, 7353–7365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Peng, Y.; Guo, G.; Shu, B.; Liu, D.; Su, P.; Zhang, X.; Gao, F. Spinal CX3CL1/CX3CR1 May Not Directly Participate in the Development of Morphine Tolerance in Rats. Neurochem. Res. 2017, 42, 3254–3267. [Google Scholar] [CrossRef]
  217. 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] [Green Version]
  218. Cataldo, G.; Erb, S.J.; Lunzer, M.M.; Luong, N.; Akgün, E.; Portoghese, P.S.; Olson, J.K.; Simone, D.A. The bivalent ligand MCC22 potently attenuates hyperalgesia in a mouse model of cisplatin-evoked neuropathic pain without tolerance or reward. Neuropharmacology 2019, 158, 107598. [Google Scholar] [CrossRef]
  219. Cataldo, G.; Lunzer, M.M.; Olson, J.K.; Akgün, E.; Belcher, J.D.; Vercellotti, G.M.; Portoghese, P.S.; Simone, D.A. Bivalent ligand MCC22 potently attenuates nociception in a murine model of sickle cell disease. Pain 2018, 159, 1382–1391. [Google Scholar] [CrossRef]
  220. Dutta, R.; Lunzer, M.M.; Auger, J.L.; Akgün, E.; Portoghese, P.S.; Binstadt, B.A. A bivalent compound targeting CCR5 and the mu opioid receptor treats inflammatory arthritis pain in mice without inducing pharmacologic tolerance. Arthritis Res. Ther. 2018, 20, 154. [Google Scholar] [CrossRef] [Green Version]
  221. Pan, H.L.; Wu, Z.Z.; Zhou, H.Y.; Chen, S.R.; Zhang, H.M.; Li, D.P. Modulation of pain transmission by G-protein-coupled receptors. Pharmacol. Ther. 2008, 117, 141–161. [Google Scholar] [CrossRef] [Green Version]
  222. Lu, Z.L.; Saldanha, J.W.; Hulme, E.C. Seven-transmembrane receptors: Crystals clarify. Trends Pharmacol. Sci. 2002, 23, 140–146. [Google Scholar] [CrossRef]
  223. Hur, E.M.; Kim, K.T. G protein-coupled receptor signalling and cross-talk: Achieving rapidity and specificity. Cell Signal. 2002, 14, 397–405. [Google Scholar] [CrossRef]
  224. Neves, S.R.; Ram, P.T.; Iyengar, R. G protein pathways. Science 2002, 296, 1636–1639. [Google Scholar] [CrossRef] [PubMed]
  225. Jeong, S.W.; Ikeda, S.R. Effect of G protein heterotrimer composition on coupling of neurotransmitter receptors to N-type Ca(2+) channel modulation in sympathetic neurons. Proc. Natl. Acad. Sci. USA 2000, 97, 907–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Leaney, J.L.; Milligan, G.; Tinker, A. The G protein alpha subunit has a key role in determining the specificity of coupling to, but not the activation of, G protein-gated inwardly rectifying K(+) channels. J. Biol. Chem. 2000, 275, 921–929. [Google Scholar] [CrossRef] [Green Version]
  227. Jensen, A.A.; Spalding, T.A. Allosteric modulation of G-protein coupled receptors. Eur. J. Pharm. Sci. 2004, 21, 407–420. [Google Scholar] [CrossRef]
  228. Gether, U. Uncovering molecular mechanisms involved in activation of G protein-coupled receptors. Endocr. Rev. 2000, 21, 90–113. [Google Scholar] [CrossRef]
  229. Chen, Y.; Mestek, A.; Liu, J.; Hurley, J.A.; Yu, L. Molecular cloning and functional expression of a mu-opioid receptor from rat brain. Mol. Pharmacol. 1993, 44, 8–12. [Google Scholar] [CrossRef]
  230. Chen, Y.; Mestek, A.; Liu, J.; Yu, L. Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the mu and delta opioid receptors. Biochem. J. 1993, 295, 625–628. [Google Scholar] [CrossRef]
  231. Kieffer, B.L.; Befort, K.; Gavériaux-Ruff, C.; Hirth, C.G. The delta-opioid receptor: Isolation of a cDNA by expression cloning and pharmacological characterization. Proc. Natl. Acad. Sci. USA 1994, 91, 1193. [Google Scholar] [CrossRef] [Green Version]
  232. Arai, H.; Charo, I.F. Differential regulation of G-protein-mediated signaling by chemokine receptors. J. Biol. Chem. 1996, 271, 21814–21819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Myers, S.J.; Wong, L.M.; Charo, I.F. Signal transduction and ligand specificity of the human monocyte chemoattractant protein-1 receptor in transfected embryonic kidney cells. J. Biol. Chem. 1995, 270, 5786–5792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Wu, D.; LaRosa, G.J.; Simon, M.I. G protein-coupled signal transduction pathways for interleukin-8. Science 1993, 261, 101–103. [Google Scholar] [CrossRef] [PubMed]
  235. Zheng, J.; Ghorpade, A.; Niemann, D.; Cotter, R.L.; Thylin, M.R.; Epstein, L.; Swartz, J.M.; Shepard, R.B.; Liu, X.; Nukuna, A.; et al. Lymphotropic virions affect chemokine receptor-mediated neural signaling and apoptosis: Implications for human immunodeficiency virus type 1-associated dementia. J. Virol. 1999, 73, 8256–8267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Dupré, D.J.; Robitaille, M.; Rebois, R.V.; Hébert, T.E. The role of Gbetagamma subunits in the organization, assembly, and function of GPCR signaling complexes. Annu. Rev. Pharmacol. Toxicol. 2009, 49, 31–56. [Google Scholar] [CrossRef] [Green Version]
  237. Bajetto, A.; Barbero, S.; Bonavia, R.; Piccioli, P.; Pirani, P.; Florio, T.; Schettini, G. Stromal cell-derived factor-1alpha induces astrocyte proliferation through the activation of extracellular signal-regulated kinases 1/2 pathway. J. Neurochem. 2001, 77, 1226–1236. [Google Scholar] [CrossRef]
  238. Li, L.-Y.; Chang, K.-J. The stimulatory effect of opioids on mitogen-activated protein kinase in Chinese hamster ovary cells transfected to express mu-opioid receptors. Mol. Pharmacol. 1996, 50, 599–602. [Google Scholar]
  239. Zhang, Z.; Xin, S.M.; Wu, G.X.; Zhang, W.B.; Ma, L.; Pei, G. Endogenous delta-opioid and ORL1 receptors couple to phosphorylation and activation of p38 MAPK in NG108-15 cells and this is regulated by protein kinase A and protein kinase C. J. Neurochem. 1999, 73, 1502–1509. [Google Scholar] [CrossRef]
  240. Wang, D.; Richmond, A. Nuclear factor-kappa B activation by the CXC chemokine melanoma growth-stimulatory activity/growth-regulated protein involves the MEKK1/p38 mitogen-activated protein kinase pathway. J. Biol. Chem. 2001, 276, 3650–3659. [Google Scholar] [CrossRef] [Green Version]
  241. Tilton, B.; Ho, L.; Oberlin, E.; Loetscher, P.; Baleux, F.; Clark-Lewis, I.; Thelen, M. Signal transduction by CXC chemokine receptor 4. Stromal cell-derived factor 1 stimulates prolonged protein kinase B and extracellular signal-regulated kinase 2 activation in T lymphocytes. J. Exp. Med. 2000, 192, 313–324. [Google Scholar] [CrossRef]
  242. Sotsios, Y.; Whittaker, G.C.; Westwick, J.; Ward, S.G. The CXC chemokine stromal cell-derived factor activates a Gi-coupled phosphoinositide 3-kinase in T lymphocytes. J. Immunol. 1999, 163, 5954–5963. [Google Scholar]
  243. Meucci, O.; Fatatis, A.; Simen, A.A.; Miller, R.J. Expression of CX3CR1 chemokine receptors on neurons and their role in neuronal survival. Proc. Natl. Acad. Sci. USA 2000, 97, 8075–8080. [Google Scholar] [CrossRef] [Green Version]
  244. Reddy, S.A.; Huang, J.H.; Liao, W.S. Phosphatidylinositol 3-kinase in interleukin 1 signaling. Physical interaction with the interleukin 1 receptor and requirement in NFkappaB and AP-1 activation. J. Biol. Chem. 1997, 272, 29167–29173. [Google Scholar] [CrossRef] [Green Version]
  245. Reddy, S.A.; Huang, J.H.; Liao, W.S. Phosphatidylinositol 3-kinase as a mediator of TNF-induced NF-kappa B activation. J. Immunol. 2000, 164, 1355–1363. [Google Scholar] [CrossRef] [Green Version]
  246. Eisenstein, T.K.; Chen, X.; Inan, S.; Meissler, J.J.; Tallarida, C.S.; Geller, E.B.; Rawls, S.M.; Cowan, A.; Adler, M.W. Chemokine Receptor Antagonists in Combination with Morphine as a Novel Strategy for Opioid Dose Reduction in Pain Management. Mil. Med. 2020, 185, 130–135. [Google Scholar] [CrossRef] [Green Version]
  247. Bogacka, J.; Popiolek-Barczyk, K.; Pawlik, K.; Ciechanowska, A.; Makuch, W.; Rojewska, E.; Dobrogowski, J.; Przeklasa-Muszynska, A.; Mika, J. CCR4 antagonist (C021) influences the level of nociceptive factors and enhances the analgesic potency of morphine in a rat model of neuropathic pain. Eur. J. Pharmacol. 2020, 880, 173166. [Google Scholar] [CrossRef]
  248. Kwiatkowski, K.; Pawlik, K.; Ciapała, K.; Piotrowska, A.; Makuch, W.; Mika, J. Bidirectional Action of Cenicriviroc, a CCR2/CCR5 Antagonist, Results in Alleviation of Pain-Related Behaviors and Potentiation of Opioid Analgesia in Rats With Peripheral Neuropathy. Front. Immunol. 2020, 11, 615327. [Google Scholar] [CrossRef]
  249. 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. Neuropsychopharmacol. Biol. Psychiatry 2016, 64, 68–78. [Google Scholar] [CrossRef]
  250. Pawlik, K.; Piotrowska, A.; Kwiatkowski, K.; Ciapała, K.; Popiolek-Barczyk, K.; Makuch, W.; Mika, J. The blockade of CC chemokine receptor type 1 influences the level of nociceptive factors and enhances opioid analgesic potency in a rat model of neuropathic pain. Immunology 2020, 159, 413–428. [Google Scholar] [CrossRef]
  251. Zychowska, M.; Rojewska, E.; Piotrowska, A.; Kreiner, G.; Nalepa, I.; Mika, J. Spinal CCL1/CCR8 signaling interplay as a potential therapeutic target—Evidence from a mouse diabetic neuropathy model. Int. Immunopharmacol. 2017, 52, 261–271. [Google Scholar] [CrossRef]
  252. Kwiatkowski, K.; Popiolek-Barczyk, K.; Piotrowska, A.; Rojewska, E.; Ciapała, K.; Makuch, W.; Mika, J. Chemokines CCL2 and CCL7, but not CCL12, play a significant role in the development of pain-related behavior and opioid-induced analgesia. Cytokine 2019, 119, 202–213. [Google Scholar] [CrossRef] [PubMed]
  253. Kwiatkowski, K.; Piotrowska, A.; Rojewska, E.; Makuch, W.; Mika, J. The RS504393 Influences the Level of Nociceptive Factors and Enhances Opioid Analgesic Potency in Neuropathic Rats. J. Neuroimmune Pharmacol. 2017, 12, 402–419. [Google Scholar] [CrossRef] [Green Version]
  254. 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]
  255. Bogacka, J.; Ciapała, K.; Pawlik, K.; Kwiatkowski, K.; Dobrogowski, J.; Przeklasa-Muszynska, A.; Mika, J. CCR4 Antagonist (C021) Administration Diminishes Hypersensitivity and Enhances the Analgesic Potency of Morphine and Buprenorphine in a Mouse Model of Neuropathic Pain. Front. Immunol. 2020, 11, 1241. [Google Scholar] [CrossRef]
  256. Rojewska, E.; Zychowska, M.; Piotrowska, A.; Kreiner, G.; Nalepa, I.; Mika, J. Involvement of Macrophage Inflammatory Protein-1 Family Members in the Development of Diabetic Neuropathy and Their Contribution to Effectiveness of Morphine. Front. Immunol. 2018, 9, 494. [Google Scholar] [CrossRef] [Green Version]
  257. Pawlik, K.; Ciechanowska, A.; Ciapała, K.; Rojewska, E.; Makuch, W.; Mika, J. Blockade of CC Chemokine Receptor Type 3 Diminishes Pain and Enhances Opioid Analgesic Potency in a Model of Neuropathic Pain. Front. Immunol. 2021, 12, 781310. [Google Scholar] [CrossRef]
  258. Bu, H.L.; Xia, Y.Z.; Liu, P.M.; Guo, H.M.; Yuan, C.; Fan, X.C.; Huang, C.; Wen, Y.Y.; Kong, C.L.; Wang, T.; et al. The Roles of Chemokine CXCL13 in the Development of Bone Cancer Pain and the Regulation of Morphine Analgesia in Rats. Neuroscience 2019, 406, 62–72. [Google Scholar] [CrossRef]
  259. Guo, G.; Peng, Y.; Xiong, B.; Liu, D.; Bu, H.; Tian, X.; Yang, H.; Wu, Z.; Cao, F.; Gao, F. Involvement of chemokine CXCL11 in the development of morphine tolerance in rats with cancer-induced bone pain. J. Neurochem. 2017, 141, 553–564. [Google Scholar] [CrossRef] [Green Version]
  260. Piotrowska, A.; Rojewska, E.; Pawlik, K.; Kreiner, G.; Ciechanowska, A.; Makuch, W.; Zychowska, M.; Mika, J. Pharmacological blockade of CXCR3 by (±)-NBI-74330 reduces neuropathic pain and enhances opioid effectiveness—Evidence from in vivo and in vitro studies. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3418–3437. [Google Scholar] [CrossRef]
  261. Wang, S.F.; Dong, C.G.; Yang, X.; Yin, J.J. Upregulation of (C-X-C motif) Ligand 13 (CXCL13) Attenuates Morphine Analgesia in Rats with Cancer-Induced Bone Pain. Med. Sci. Monit. 2016, 22, 4612–4622. [Google Scholar] [CrossRef] [Green Version]
  262. Ye, D.; Bu, H.; Guo, G.; Shu, B.; Wang, W.; Guan, X.; Yang, H.; Tian, X.; Xiang, H.; Gao, F. Activation of CXCL10/CXCR3 signaling attenuates morphine analgesia: Involvement of Gi protein. J. Mol. Neurosci. 2014, 53, 571–579. [Google Scholar] [CrossRef] [PubMed]
  263. Reverte, I.; Pujol, A.; Domingo, J.L.; Colomina, M.T. Thyroid hormones and fear learning but not anxiety are affected in adult apoE transgenic mice exposed postnatally to decabromodiphenyl ether (BDE-209). Physiol. Behav. 2014, 133, 81–91. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Scheme of the multidisciplinary approach for the management of neuropathic pain. Patient interview, physical and clinical tests and screening questionnaires-Leeds Assessment of Neuropathic Symptoms and Signs (LANSS) [14], Neuropathic Pain Questionnaire (NPQ) [15], painDETECT [16], Douleur Neuropathique en 4 Questions (DN4) [17], ID PAIN [18]—are the main steps of a comprehensive diagnosis [8] required to characterize neurological lesions, determine the presence of comorbidities, identify typical neuropathic symptoms and signs [19] and also assess the emotional, social, and economic impact of NP on patients’ lives [20]. Focusing on clinical symptoms rather than causative events/factors [2,10], NP treatment currently includes both pharmacological and non-pharmacological (interventional, physical, and psychological) options [8,9,10,21,22] whose efficacy and safety are constantly monitored with the following assessment questionnaires: Neuropathic Pain Scale (NPS) [23], Neuropathic Pain Symptoms Inventory (NPSI) [24], Pain Quality Assessment Scale (PQAS) [25], and the McGill short-form questionnaire 2 [26]. Among pharmacological options, according to the Grading of Recommendation, Assessment, Development and Evaluation (GRADE) system, gabapentinoids, tricyclic antidepressants (TCAs), serotonin-norepinephrine reuptake inhibitors (SNRIs), and anticonvulsants (sodium ion channel blockers) are strongly recommended for use and proposed by international guidelines as first-line drugs; weak opioids, lidocaine patches, and 8% capsaicin patches are weakly recommended for use and classified as second-line drugs; strong opioids and botulinum toxin type A have only weak GRADE recommendation and are relegate among third-line drugs.
Figure 1. Scheme of the multidisciplinary approach for the management of neuropathic pain. Patient interview, physical and clinical tests and screening questionnaires-Leeds Assessment of Neuropathic Symptoms and Signs (LANSS) [14], Neuropathic Pain Questionnaire (NPQ) [15], painDETECT [16], Douleur Neuropathique en 4 Questions (DN4) [17], ID PAIN [18]—are the main steps of a comprehensive diagnosis [8] required to characterize neurological lesions, determine the presence of comorbidities, identify typical neuropathic symptoms and signs [19] and also assess the emotional, social, and economic impact of NP on patients’ lives [20]. Focusing on clinical symptoms rather than causative events/factors [2,10], NP treatment currently includes both pharmacological and non-pharmacological (interventional, physical, and psychological) options [8,9,10,21,22] whose efficacy and safety are constantly monitored with the following assessment questionnaires: Neuropathic Pain Scale (NPS) [23], Neuropathic Pain Symptoms Inventory (NPSI) [24], Pain Quality Assessment Scale (PQAS) [25], and the McGill short-form questionnaire 2 [26]. Among pharmacological options, according to the Grading of Recommendation, Assessment, Development and Evaluation (GRADE) system, gabapentinoids, tricyclic antidepressants (TCAs), serotonin-norepinephrine reuptake inhibitors (SNRIs), and anticonvulsants (sodium ion channel blockers) are strongly recommended for use and proposed by international guidelines as first-line drugs; weak opioids, lidocaine patches, and 8% capsaicin patches are weakly recommended for use and classified as second-line drugs; strong opioids and botulinum toxin type A have only weak GRADE recommendation and are relegate among third-line drugs.
Life 12 00398 g001
Figure 2. Involvement of chemokines and chemokine receptors in opioid-induced analgesia in NP. (A) Neuropathic pain conditions increase the production and release of a myriad of chemokines, whose binding to their specific receptors on neurons, astrocytes, and microglia decreases the nociceptive pain threshold and increases glial activation. Additionally, chronic μ-agonists administration further increases the chemokines and chemokine receptor expression and enhances the astrocyte and microglia reactivity in CNS. The neuronal activation of chemokine receptors by their specific ligands is followed by complex biochemical events that lead to heterodimer formation or heterologous desensitization between chemokine and opioid receptors, which lead to neuronal sensitization and reduce the opioid-induced analgesia. (B) The administration of chemokine-neutralizing specific antibodies or chemokine receptor specific antagonists reduces pain-related behaviors and glial cells activation and improve opioid efficacy in neuropathic conditions.
Figure 2. Involvement of chemokines and chemokine receptors in opioid-induced analgesia in NP. (A) Neuropathic pain conditions increase the production and release of a myriad of chemokines, whose binding to their specific receptors on neurons, astrocytes, and microglia decreases the nociceptive pain threshold and increases glial activation. Additionally, chronic μ-agonists administration further increases the chemokines and chemokine receptor expression and enhances the astrocyte and microglia reactivity in CNS. The neuronal activation of chemokine receptors by their specific ligands is followed by complex biochemical events that lead to heterodimer formation or heterologous desensitization between chemokine and opioid receptors, which lead to neuronal sensitization and reduce the opioid-induced analgesia. (B) The administration of chemokine-neutralizing specific antibodies or chemokine receptor specific antagonists reduces pain-related behaviors and glial cells activation and improve opioid efficacy in neuropathic conditions.
Life 12 00398 g002
Table 1. Human neuropathic pain and its corresponding animal model. Partial list of animal models used to mimic human neuropathic pain.
Table 1. Human neuropathic pain and its corresponding animal model. Partial list of animal models used to mimic human neuropathic pain.
CategoriesHuman
Neuropathic Pain
Type of Injury in Animal ModelsSpecies
Central pain
models
Thalamic syndrome
(stroke in the
thalamus)
Collagenase injection in
ventral posterolateral
nucleus of the thalamus [42]
Rats
Mechanical injury
to the spinal cord
Contusion or constant weight dropped
over the spinal cord [43]
Intraspinal injections of excitotoxins
or excitatory amino acids [44,45]
Injection of Erythrosin B (photosensitizing dye) through
the tail vein followed by surgical exposure
of vertebrae to an argon ion laser [46,47]
Rats, Mice
Hemisection [48]Rats
Peripheral pain modelsComplete nerve
lesion
Complete transection of sciatic nerve [49]
Brachial plexus avulsion [50,51]
Caudal trunk resection [52]
Rats, Mice
Tibial and sural nerve transection [53]Rats
Partial nerve
lesion
Sciatic nerve chronic constriction injury [54]
Partial sciatic nerve ligation [55]
Spinal nerve ligation [56,57]
Axotomy of tibial nerve and/or common
peroneal nerves and/or sural nerve [58,59]
Partial injury of the saphenous nerve [60,61]
Injection of Zymosan, HMG, and TNFα in the sciatic nerve [62,63,64,65]
Implanting of polyethylene cuff around the common branch of the sciatic nerve [66,67]
Rats, Mice
Freezing of the sciatic nerve with a cryoprobe [68]
Laser irradiation [69]
Rats
Ligation of common peroneal nerve [70]Mice
Trigeminal
neuralgia
Chronic constriction injury of
infraorbital trigeminal branch [71]
Rats
Partial ligation of the infraorbital trigeminal branch [72]Mice
Orofacial painInjection of formalin into the temporomandibular joints [73]
Subcutaneous injection of carrageenan
over the right maxilla [74]
Rats, Mice
Disease-induced pain modelsMultiple sclerosisInduction of experimental autoimmune encephalomyelitis by
immunization with myelin oligodendrocyte glycoprotein [75]
Intracerebral inoculation of Theiler’s murine
encephalomyelitis virus [76]
Mice
Postherpetic
neuralgia
Subcutaneous injection of cells infected with
varicella zoster virus in the foot [77,78]
Injection of herpes simplex virus in the skin
of the hind paw [79]
Administration of resiniferotoxin, TRPV-1 agonist, for depletion of capsaicin-sensitive afferents [80]
Rats, Mice
HIV-associated
sensory neuropathy
HIV-protein gp120 delivery in sciatic nerve [81]Rats
Transgenic animals expressing HIV-protein gp120 under a GFAP promoter [82]Mice
Cancer painDirect inoculation of compatible murine cancer cells [83,84]
Direct inoculation of tumor cells [85,86]
Rats, Mice
DiabetesAdministration of the pancreatic B-cell toxins
streptozotocin [87,88]
Administration of alloxan [89]
Transgenic animals of type I and II diabetes [90,91,92,93]
Rats, Mice
Drug-induced
neuropathy models
Anti-cancer
agents-induced
neuropathy
Administration of vincristine, cisplatin,
oxaliplatin, or taxanes [94,95,96,97]
Rats, Mice, Guinea pigs
Anti-HIV
drugs-induced
neuropathy
Administration of 2,3-dideoxycytidine [98]Rabbits
Administration of didanosine [99]Rats
Inherited-induced pain modelsSpontaneous
neuropathy
Mutations of Trembler (Tr) and Trembler-J (Tr-J) in the
PMP22 myelin gene [100]
PMP22-transgenic animals [101,102]
Mutations encoding for the myelin components
P0 and connexin 32 [103]
Mice
Table 2. Novel target strategies that may increase the opioid analgesic efficacy in neuropathic pain.
Table 2. Novel target strategies that may increase the opioid analgesic efficacy in neuropathic pain.
Chemokine/
Chemokine
Receptors Axis
TargetChemokine/
Chemokine
Receptor
Inhibitors
Effects on Opioid Efficacy under Neuropathic Pain
CCL1/CCR8CCL1Neutralizing
antibody
↑ Analgesic effects of morphine and buprenorphine (STZ, mice) [251]
CCL2/CCR2- CCR4CCL2Neutralizing
antibody
↑ Analgesic effects of morphine and buprenorphine (CCI, mice) [252]
CCR2RS504393
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, rats) [253]
Cenicriviroc
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, rats) [248]
CCR4C021
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, mice) and delays the development of morphine-induced tolerance [255]
CCL3- CCL4- CCL5/CCR5CCR5Maraviroc
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, rats) [249]
Cenicriviroc
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, rats) [248]
CCL3- CCL9/CCR1CCL3Neutralizing
antibody
↑ Analgesic effects of morphine (STZ, mice) [256]
CCL9Neutralizing
antibody
↑ Analgesic effects of morphine (STZ, mice) [256]
CCR1J113863
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (STZ, mice; CCI, rats) [250,256]
CCL7/CCR2CCL7Neutralizing
antibody
↑ Analgesic effects of morphine and buprenorphine (CCI, mice) [252]
CCL7- CCL11/CCR3CCR3SB328437
(antagonist)
↑ Analgesic effects of morphine and buprenorphine (CCI, rats) [257]
CXCL4- CXCL9- CXCL10- CXCL11- CCL21/
CXCR3
CXCL10Neutralizing
antibody
↑ Analgesic effects of morphine (BCP, rats) [262]
CXCL11Neutralizing
antibody
↑ Analgesic effects of morphine (BCP, rats) and attenuates morphine-induced tolerance [259]
CXCR3NBI-74330
(antagonist)
↓ Levels of CXCL4, CXCL9, CXCL10, CXCL11 and CCL21 in DRG and spinal cord (CCI, rats)
↑ Analgesic effects of morphine but not of buprenorphine [260]
CXCL13/
CXCR5
CXCR13Neutralizing
antibody
↑ Analgesic effects of morphine (BCP, rats) and prevents the development of morphine-induced tolerance [261]
CXCR5siRNA↑ Analgesic effects of morphine (BCP, rats) [258]
Abbreviations: BPC-bone cancer pain; CCI-chronic constriction injury; STZ-streptozotocin diabetic neuropathy; siRNA-small interfering RNA.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vincenzi, M.; Milella, M.S.; D’Ottavio, G.; Caprioli, D.; Reverte, I.; Maftei, D. Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain. Life 2022, 12, 398. https://doi.org/10.3390/life12030398

AMA Style

Vincenzi M, Milella MS, D’Ottavio G, Caprioli D, Reverte I, Maftei D. Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain. Life. 2022; 12(3):398. https://doi.org/10.3390/life12030398

Chicago/Turabian Style

Vincenzi, Martina, Michele Stanislaw Milella, Ginevra D’Ottavio, Daniele Caprioli, Ingrid Reverte, and Daniela Maftei. 2022. "Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain" Life 12, no. 3: 398. https://doi.org/10.3390/life12030398

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