Next Article in Journal
Heparan Sulfate Biosynthetic System Is Inhibited in Human Glioma Due to EXT1/2 and HS6ST1/2 Down-Regulation
Next Article in Special Issue
IFN-β: A Contentious Player in Host–Pathogen Interaction in Tuberculosis
Previous Article in Journal
Relation among Aromatase P450 and Tumoral Growth in Human Prolactinomas
Previous Article in Special Issue
Time Course of the Phenotype of Blood and Bone Marrow Monocytes and Macrophages in the Lung after Cigarette Smoke Exposure In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages

Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(11), 2296; https://doi.org/10.3390/ijms18112296
Submission received: 19 October 2017 / Revised: 27 October 2017 / Accepted: 31 October 2017 / Published: 1 November 2017
(This article belongs to the Special Issue Macrophages in Inflammation)

Abstract

:
Neuropathic pain can have a major effect on quality of life but current therapies are often inadequate. Growing evidence suggests that neuropathic pain induced by nerve damage is caused by chronic inflammation. Upon nerve injury, damaged cells secrete pro-inflammatory molecules that activate cells in the surrounding tissue and recruit circulating leukocytes to the site of injury. Among these, the most abundant cell type is macrophages, which produce several key molecules involved in pain enhancement, including cytokines and chemokines. Given their central role in the regulation of peripheral sensitization, macrophage-derived cytokines and chemokines could be useful targets for the development of novel therapeutics. Inhibition of key pro-inflammatory cytokines and chemokines prevents neuroinflammation and neuropathic pain; moreover, recent studies have demonstrated the effectiveness of pharmacological inhibition of inflammatory (M1) macrophages. Nicotinic acetylcholine receptor ligands and T helper type 2 cytokines that reduce M1 macrophages are able to relieve neuropathic pain. Future translational studies in non-human primates will be crucial for determining the regulatory mechanisms underlying neuroinflammation-associated neuropathic pain. In turn, this knowledge will assist in the development of novel pharmacotherapies targeting macrophage-driven neuroinflammation for the treatment of intractable neuropathic pain.

Graphical Abstract

1. Introduction

Neuropathic pain elicited by damage to or dysfunction of the sensory nervous system severely affects quality of life and is associated with a high economic cost for both the individual and society [1,2,3]. The symptoms of neuropathic pain are sensory hypersensitivity defined by spontaneous pain, hyperalgesia, and allodynia [4,5,6] resistant to standard analgesics [7,8]. Neuropathic pain can be caused by not only physical lesions (e.g., traumatic nerve injury and spinal cord injury) but also other reasons, such as diabetes, chemotherapy, and viral infection [9,10,11], and senior people have a high risk of suffering neuropathic pain [12,13]. It is difficult to treat all types of hypersensitivity with currently available medications, and many patients with neuropathic pain do not receive appropriate treatment [14,15,16]. Thus, there is an urgent need for evidence-based development of novel pharmacotherapies for neuropathic pain.
A number of experimental animal models [17,18,19,20] have contributed to our understanding of the key components of neuropathic pain. In particular, recent studies have suggested that interactions between the nervous and immune systems trigger chronic neuroinflammation resulting in aberrant sensory processing and neuropathic pain [17,21,22]. Upon nerve injury, several cell types, including damaged neurons, demyelinated Schwann cells, and tissue-resident macrophages, produce soluble inflammatory cytokines, chemokines, and damage-associated molecular patterns (DAMPs) that activate surrounding cells [23,24,25,26,27,28] and recruit circulating leukocytes, such as monocytes/macrophages, neutrophils, and lymphocytes, into the site of injury [22,29,30]. Numerous pro-inflammatory cytokines (e.g., interleukin-1β (IL-1β), IL-6, and tumor necrosis factor α (TNFα)) and chemokines (e.g., CC-chemokine ligand 2 (CCL2, also known as monocyte chemoattractant protein-1), CCL3, and CCL4) are released by the infiltrating leukocytes, directly sensitize nociceptors, and alter the processing of nociceptive information by sensory (dorsal root ganglion, DRG) neurons [31,32,33,34]. Several studies using different rodent models have shown that inhibition of these leukocyte functions prevents the pathogenesis of neuropathic pain [29,35,36,37]. Macrophages have especially significant functions in regulating neuroinflammation; consequently, they are considered to be a common peripheral regulator of neuropathic pain [25,30,38].
The contribution of macrophages to neuropathic pain was mainly uncovered in studies of rodents in which macrophages were depleted by toxin treatment or genetic ablation, which may have had other effects on homeostasis [36,37]. Thus, there is a significant knowledge gap between the rodent studies and the human situation that must be filled before these findings can be translated to the clinic. Nonetheless, it may be possible to develop novel therapeutics for neuropathic pain by pharmacological targeting of immunoregulatory systems in animals without genetic abnormalities. In this review, we highlight our current understanding of how macrophages mediate the pathogenesis of neuropathic pain, and how macrophage activity could be modulated to develop new pharmacological approaches for the relief of intractable neuropathic pain.

2. Peripheral and Central Sensitization

It is well known that pain sensation is processed by a discriminative set of primary afferent neurons [39,40]. Unmyelinated C-fibers and thinly myelinated Aδ fibers act as nociceptors, while myelinated Aβ fibers are tactile sensors [41,42]. Noxious stimuli such as heat, cold, pressure, and chemicals are converted to electrical activity by distinct cation channels (e.g., transient receptor potential (TRP) channels and sodium channels) that elicit action potentials [43,44,45,46]. Primary afferent neurons producing glutamate or neuropeptides transmit peripheral information to secondary neurons in the spinal dorsal horn [39,40]. During neuropathic pain, the expression and sensitivity of these channels become dysregulated and elicit ectopic activity of nociceptive DRG neurons [46,47,48]. Despite the complexity of the underlying mechanisms, the close relationship between ectopic activity and pro-inflammatory mediators has been noted in several studies [21,22,49,50]. Because many nociceptive DRG neurons express pro-inflammatory cytokine and chemokine receptors that are upregulated after nerve injury, pro-inflammatory molecules can directly sensitize nociceptors, such as TRP channels, in C-fibers leading to hypersensitivity. For example, IL-1β, TNFα, IL-6, CCL2, and CCL3 are well-known enhancers of nociceptor activity [31,32,33,34,51]. Thus, long-lasting neuroinflammation resulting from upregulation of inflammatory molecules by damaged tissue and infiltrating leukocytes can contribute to the ectopic discharge of sensory neurons, resulting in peripheral sensitization.
Prolonged abnormal transmission of pain signaling into the spinal dorsal horn due to peripheral sensitization triggers central sensitization [6,52,53], characterized by increased excitability of pain-processing neurons and activation of glial cells (microglia and astrocytes) [17,54,55,56]. These glial cells have been the focus of increasing attention in the past few decades, and their critical contribution to spinal neuroinflammation underlying neuropathic pain is now well characterized [29,57,58,59,60]. Microglia and astrocytes are activated by several neurotransmitters derived from primary afferent neurons, such as cytokines, chemokines, and nucleotides. Activation of glial cells induces a variety of pro-inflammatory factors that directly or indirectly sensitize pain-processing neurons in the spinal dorsal horn [17,54,55,56,61]. Similar to the peripheral response, typical inflammatory cytokines (IL-1β, TNFα, IL-6), chemokines (CCL2 and CCL3), and growth factors are upregulated in the dorsal horn after nerve injury, and inhibition of these molecules reverses neuropathic pain [62,63,64,65]. These pain-facilitating molecules function to sensitize ionotropic glutamate receptors such as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor and N-methyl-d-aspartate (NMDA) receptors [39,55,56,66]. Because AMPA and NMDA receptors play central roles in pain processing in the spinal cord, modulation of their sensitivity by pro-inflammatory mediators derived from activated glial cells is also important for the pathogenesis of neuropathic pain.

3. Accumulation of Macrophages in Injured Nerves

Macrophages are a fundamental component of innate immunity and play pivotal roles in the regulation of inflammatory responses and in the host defense against pathogens [67,68]. Upon tissue injury or infection, chemotactic factors, such as chemokines, recruit circulating monocytes to the site of damage. The monocytes then differentiate into macrophages that orchestrate the inflammatory response through cytokine and chemokine production to repair damaged tissues and/or eliminate the source of infection [67,68,69]. In addition, the majority of tissues contain resident macrophages that contribute to the maintenance of tissue homeostasis and the resolution of localized inflammation [70,71,72]. The variety of receptors expressed by tissue-resident macrophages enables their rapid activation by pro-inflammatory cytokines, chemokines, and DAMPs (e.g., high mobility group box 1, HMGB1) produced by damaged cells [71,73,74,75]. Following nerve injury, resident macrophages and Schwann cells synergistically initiate inflammatory responses and elicit long-lasting neuroinflammation through the recruitment of circulating leukocytes to the site of injury [25,29,76].
Neutrophils and lymphocytes also infiltrate into the sites of peripheral nerve injury in rodents [37,77,78,79,80], and depletion of either cell types using experimental drugs or genetic ablation significantly reduces neuropathic pain resulting from neuroinflammation [35,36,37,81,82]. Figure 1 shows a typical analysis of infiltrating macrophages, neutrophils, and T lymphocytes in the injured and contralateral sciatic nerves of mice 1 week after partial sciatic nerve ligation by flow cytometry using well-characterized marker of each leukocyte. An increased number of CD11b+ cells was seen in the injured nerve compared with the intact nerve. This population contains F4/80+ macrophages, while the F4/80 population included Ly6G+ neutrophils and CD3+ T lymphocytes. The abundance of each cell type is clearly higher in the injured than in the intact nerve. Importantly, macrophages make up the largest population of infiltrating cells, supporting the notion that they may play a central role in the regulation of neuroinflammation.
Macrophage accumulation is consistently observed in rodent models of experimental neuropathic pain [38], including that resulting from physical nerve damage (e.g., partial sciatic nerve ligation and chronic constriction injury) [83,84,85], diabetes, and chemotherapy (paclitaxel and vincristine) [86,87,88,89]. Notably, targeted inhibition or depletion of macrophages prevents neuroinflammation and pain hypersensitivity in these neuropathic pain models [36,88,90,91]. Collectively, these studies strongly support a critical role for macrophages in peripheral neuroinflammation leading to neuropathic pain.

4. Macrophage Polarization and Neuroinflammation

Macrophages are remarkably plastic and can be polarized towards some functionally distinct phenotypes [67,68,92]. Molecules known as “pathogen-associated molecular patterns,” such as bacterial lipopolysaccharides (LPS), and interferon-γ, a cytokine produced by T helper type 1 (Th1) cells, induce the differentiation of classically activated (M1) macrophages that facilitate and exacerbate inflammation [93,94]. In contrast, Th2 cytokines (IL-4 and IL-13) promote differentiation into alternatively activated (M2) macrophages, which suppress inflammation and promote its resolution [75,95]. M1 macrophages are characterized by high expression of pro-inflammatory cytokines such as IL-1β, IL-6, TNFα, and receptors such as Toll-like receptor 4 (TLR4, the LPS receptor), major histocompatibility class II (MHC II), and CD86 [75,93,94]. M1 macrophage-dominant molecules are preferentially upregulated through signaling via signal transducer and activator of transcription 1 (STAT1) and interferon regulatory factor 5 (IRF5) pathways [75,94,96,97]. In contrast, M2 macrophages abundantly express the anti-inflammatory cytokine IL-10 and the scavenger receptors CD163 and CD206 (mannose receptor C type 1) [75,94]. Unlike M1 macrophages, the dominant signaling pathways are via STAT6 and IRF4 in M2 macrophages [97,98]. Chemokine expression patterns also differ between M1 and M2 macrophages [92,93,99,100]; thus, CCL2, CCL3, CCL4 and CCL5 are highly produced by M1 macrophages, whereas CCL18, CCL22 and CCL24 are abundantly expressed by M2 macrophages [99,100,101,102].
The soluble mediators produced by M1 and M2 macrophages are functionally antagonistic [94,97], and the balance between M1 and M2 macrophages is therefore closely related to the progression of inflammatory disorders, including neuropathic pain [103,104]. Prolonged activation of M1 macrophages has been reported to drive non-resolving neuroinflammation that underlies the pathogenesis of neuropathic pain [103,104,105]. Under normal conditions, tissue-resident macrophages are predominantly M2-like and function to protect the peripheral nervous system from damage and to maintain tissue homeostasis [70,72]. However, when the balance tilts in favor of M1 macrophages at the population level, the upregulated expression of M1-dominant molecules has diverse effects on the surrounding cells [75,92,93]. In addition, inflammatory chemokine secretion recruits circulating leukocytes into the inflamed tissue, resulting in long-lasting inflammation [67,69,106]. Since macrophage polarization and activation can be diverse in response to environmental cues [107,108], inhibition of M1 macrophage differentiation has been proposed as a pharmacotherapy for the treatment of neuroinflammatory diseases such as neuropathic pain.

5. Roles of Cytokines and Chemokines in Neuropathic Pain

Cytokines and chemokines regulate many aspects of the immune response, including cell-to-cell interactions [109,110,111,112]. IL-1β, IL-6 and TNFα are well-characterized M1 macrophage-dominant cytokines and enhancers of inflammation [92,99,101]. Emerging evidence suggests that these cytokines are upregulated in injured nerves and may contribute significantly to neuropathic pain [17,21,29,113,114]. These cytokines are also produced by Schwann cells and infiltrating leukocytes, suggesting indicating that interactions between these cell types plays a role in injury-induced neuroinflammation [22,25,30,49]. Indeed, upregulation of pro-inflammatory cytokines has been observed in diverse neuropathic pain models (e.g., nerve ligation, diabetes, and chemotherapeutics) [17,38,56,115], and inhibition of their functions using neutralizing antibodies, receptor antagonists, intracellular signaling pathway inhibitors, or genetic ablation significantly reduce hypersensitivity associated with neuroinflammation [23,29,89,116,117,118].
Chemokines are small chemotactic cytokines that also play fundamental roles in neuroinflammation [110,119,120]. These molecules recruit circulating leukocytes into the inflamed tissue and activate various cell types via engagement of cell surface receptors [69,120]. Of the four chemokine families, CC-chemokines are the most well-established regulators of neuropathic pain [30,121,122,123]. In particular, many studies have demonstrated that CCL2 is upregulated in macrophages and damaged Schwann cells following nerve injury [26,51,124,125], and pharmacological or genetic inhibition of CCL2 and/or its receptor CCR2 markedly alleviates neuropathic pain [126,127,128].
Recently, CCL3 (macrophage inflammatory protein-1α, MIP-1α) and CCL4 (MIP-1β) have been shown to be upregulated in macrophages and Schwann cells following nerve injury, and to play critical roles in orchestrating long-lasting neuroinflammation in the peripheral nervous system [23,30,123,129]. Because their receptors (CCR1 and CCR5) are also expressed on macrophages and Schwann cells, CCL3 and CCL4 can act in an autocrine fashion to drive intracellular signaling for upregulation of inflammatory molecules such as IL-1β. Accordingly, blockade of CCL3, CCL4, CCR1, or CCR5 function with neutralizing antibodies or inhibitors prevents the upregulation of IL-1β and other M1-dominant cytokines and thus reduces neuropathic pain [23,129]. CCL5 also contributes to the upregulation of M1-dominant cytokines and the recruitment of circulating monocytes, neutrophils, and T lymphocytes to the site of injury [130]. Previous reports have shown that these chemokine receptors are expressed on DRG neurons [33,34,51,131], indicating that CC-chemokines can not only activate inflammatory cells but also directly sensitize nociceptive neurons and enhance pain processing. The CXC-chemokine ligand 2 (CXCL2, also known as macrophage inflammatory protein-2, MIP-2) plays a pivotal role in generating neuropathic pain by mediating interactions between macrophages and neutrophils [35]. CXCL2 and its receptor CXCR2 are upregulated in both cell types in the environment of injured nerves [35]. Neutralizing anti-CXCL2 antibodies or CXCR2 antagonists abolish neutrophil and macrophage infiltration and the upregulation of cytokines underlying neuropathic pain [35]. Therefore, inflammatory cytokines and chemokines derived from M1 macrophages and surrounding inflammatory cells could be novel therapeutic targets for neuropathic pain associated with neuroinflammation (Figure 2). Given that pharmacological inhibitors (e.g., neutralizing antibodies and small compounds) of these key mediators are currently used for inflammatory diseases in the clinic [132,133,134,135], further investigations can facilitate developing effective medications for neuropathic pain.

6. Phenotypic Shift of Macrophages by Cytokines

Since M1 macrophages produce many of the key cytokines and chemokines underlying neuropathic pain [30,99,103,122], modulation of macrophage polarization is likely to have a major effect on the pathogenesis of neuropathic pain. Several lines of evidence indicate that IL-4 and IL-13 polarize macrophages toward an M2 phenotype by binding to their receptors, which are composed of heteromeric complexes IL-4 receptor α (IL-4Rα)/common γc chain and IL-4Rα/IL-13Rα1, respectively [136,137]. Downstream signaling through these receptor complexes is mediated by phosphorylation of STAT6 and activation of IRF4, which upregulates the expression of M2-dominant molecules [97,98]. As mentioned, the progression of inflammatory diseases is strongly influenced by the balance between M1 and M2 macrophages [94,138,139]; accordingly, treatment with Th2 cytokines can improve the severity of many diseases by inducing a phenotype shift from the M1 toward the M2 phenotype [140,141,142]. These findings suggest that pharmacological targeting of macrophage polarization using Th2 cytokines may also improve neuropathic pain.
Expression level of IL-4Rα is increased upon nerve injury, mainly due to elevated production by infiltrating macrophages [103]. Notably, addition of IL-4 to LPS-stimulated macrophages in vitro or to isolated injured nerves ex vivo decreases the production of M1-dominant molecules (IL-1β and CCL3) and increases that of M2-dominant molecules (IL-10 and CD206) through activation of STAT6 [103]. Consistent with this, administration of IL-4 to injured nerves may directly shift the phenotype of induced macrophages from M1 toward M2, resulting in the relief of neuropathic pain [103]. Moreover, IL-13 shows similar effects to IL-14, and it also suppresses neuropathic pain by shifting the balance towards M2 macrophages [104]. These recent findings substantiate previous reports demonstrating that neuropathic pain is abolished or attenuated by inhibition of M1-dominant molecules or enhancement of anti-inflammatory cytokines such as IL-10 [17,22,23,30,130,143,144]. It is important to note that IL-4 and IL-13 are beneficial not only during the early phase of nerve injury but also during the middle/late phases, which is similar to the effects of ligands of α4β2 nicotinic acetylcholine receptor (nAChR) [103,104,145]. Since these effects are independent of the pathophysiological stage, infusion of Th2 cytokines may be an attractive pharmacotherapy of neuroinflammation-induced neuropathic pain, particularly since exogenous Th2 cytokines are unlikely to cause severe adverse effects (Figure 2).

7. Inhibition of Macrophages by Nicotinic Acetylcholine Receptors

Although therapeutic agents that act on pro-inflammatory mediators have had considerable success in the clinic [146,147,148,149], the effectiveness of single molecularly targeted inhibitors is sometimes limited [150,151,152,153], implying that combination multi-targeted therapy may be necessary for successful treatment of inflammatory diseases.
The peripheral immune system is modulated by the central nervous system [146,154]. Emerging evidence indicates that vagus nerve activation can suppress inflammation through nAChR-mediated inhibition of macrophage function [155,156,157,158]. Consistent with this, nicotine treatment improves a variety of intractable inflammatory diseases in rodents [146,156,157,159,160]. nAChRs are ligand-gated cation channels consisting of homo- or hetero-pentameric complexes formed from distinct subunits [161,162]. The first subtype to be demonstrated to have inhibitory effects on macrophages was α7 [155]. Nicotine treatment can inhibit the production of inflammatory cytokines by LPS-stimulated macrophages via activation of α7 nAChR signaling. Moreover, a number of studies have reported that nicotine treatment suppresses various inflammatory diseases by inhibiting production of M1 macrophage-dominant factors such as IL-1β, TNFα, and HMGB1 [156,159,160]. In contrast, neither nicotine nor other α7 nAChR ligands affect the production of M2 macrophage-dominant molecules such as IL-10 [146,156]. Although the majority of reports focused on the anti-inflammatory property of α7 nAChR, some studies have found that the α4β2 nAChR subtype has similar effects, depending on the tissue injury or disease [163,164].
Macrophages and neutrophils that accumulate around injured nerves express both α7 and α4β2 nAChRs [145,165], and administration of nicotine to the site of injury suppresses upregulation of M1-dominant cytokines and neuropathic pain [165]. Importantly, the suppressive effects of nicotine are reversed by selective antagonism of the α4β2, but not α7, subtype, revealing the dominant role of α4β2 nAChR in the regulation of inflammatory macrophage-driven neuropathic pain [145]. Furthermore, engagement of α4β2 nAChR by either local or systemic administration of the selective ligand TC-2559 markedly reduced neuropathic pain [145]. TC-2559 treatment of LPS-stimulated macrophages suppressed the expression of IL-1β and CCL3 by M1 macrophages by inhibiting the phosphorylation of STAT3 [166], a key transcription factor for these cytokines [167,168]. The observation that α4β2 nAChR ligands are effective in reducing neuropathic pain in rodents during the early, middle, and late phases of nerve injury suggests that α4β2 nAChR expressed on macrophages could be a novel pharmacological target (Figure 2).

8. Conclusions

In this review, we have highlighted the critical roles of macrophages in regulating neuropathic pain caused by peripheral neuroinflammation. A growing body of evidence supports the contribution of infiltrating leukocytes, including macrophages, to the pathogenesis of neuropathic pain, as demonstrated in several animal models. Since macrophages play a central role in regulating peripheral sensitization, cytokines and chemokines derived from these cells are potential targets for novel therapeutics, particularly as combination therapies. Approaches that use physiological modulators of macrophage function (e.g., nAChR ligands and Th2 cytokines) may be more effective and have greater safety margins because they function as endogenous immunoregulators. Most importantly, these approaches are effective at the early, middle, and late phases of neuropathic pain after nerve injury.
Although several molecules have been shown to prevent or dramatically relieve neuropathic pain in rodent models, their efficacy has not yet been evaluated in humans. Most rodent studies have employed single experimental models, and there are major systemic and anatomical differences between rodents and primates. The evidence that macrophages contribute to the pathogenesis of neuropathic pain in humans is obviously more circumstantial because of the difficulty in performing functional studies in humans [169,170,171,172]. Among the key questions that remain are whether infiltrating leukocytes mediate the sensory abnormality, and whether neuroinflammation parallels neuropathic pain in patients. The basic components of the inflammatory response are similar in rodents and primates; however, further studies will be necessary to identify the mechanisms that regulate neuroinflammation-associated neuropathic pain in humans. Development of novel evidence-based pharmacotherapies that target macrophage-driven neuroinflammation will undoubtedly open up a new avenue for the treatment of intractable neuropathic pain.

Acknowledgments

This work was supported by Smoking Research Foundation and JSPS KAKENHI Grant Numbers JP15K10563 and JP16K08994. We thank Anne M. O’Rourke, from Edanz Group (www.edanzediting.com/ac) for editing a draft of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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]
  2. Doth, A.H.; Hansson, P.T.; Jensen, M.P.; Taylor, R.S. The burden of neuropathic pain: A systematic review and meta-analysis of health utilities. Pain 2010, 149, 338–344. [Google Scholar] [CrossRef] [PubMed]
  3. Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpaa, 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]
  4. Jensen, T.S.; Baron, R.; Haanpaa, M.; Kalso, E.; Loeser, J.D.; Rice, A.S.; Treede, R.D. A new definition of neuropathic pain. Pain 2011, 152, 2204–2205. [Google Scholar] [CrossRef] [PubMed]
  5. Jensen, T.S.; Finnerup, N.B. Allodynia and hyperalgesia in neuropathic pain: Clinical manifestations and mechanisms. Lancet Neurol. 2014, 13, 924–935. [Google Scholar] [CrossRef]
  6. Von Hehn, C.A.; Baron, R.; Woolf, C.J. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012, 73, 638–652. [Google Scholar] [CrossRef] [PubMed]
  7. Finnerup, N.B.; Sindrup, S.H.; Jensen, T.S. The evidence for pharmacological treatment of neuropathic pain. Pain 2010, 150, 573–581. [Google Scholar] [CrossRef] [PubMed]
  8. Gilron, I.; Bailey, J.M.; Tu, D.; Holden, R.R.; Weaver, D.F.; Houlden, R.L. Morphine, gabapentin, or their combination for neuropathic pain. N. Engl. J. Med. 2005, 352, 1324–1334. [Google Scholar] [CrossRef] [PubMed]
  9. Baron, R. Mechanisms of disease: Neuropathic pain—A clinical perspective. Nat. Clin. Pract. Neurol. 2006, 2, 95–106. [Google Scholar] [CrossRef] [PubMed]
  10. Baron, R.; Binder, A.; Wasner, G. Neuropathic pain: Diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol. 2010, 9, 807–819. [Google Scholar] [CrossRef]
  11. Van Hecke, O.; Austin, S.K.; Khan, R.A.; Smith, B.H.; Torrance, N. Neuropathic pain in the general population: A systematic review of epidemiological studies. Pain 2014, 155, 654–662. [Google Scholar] [CrossRef] [PubMed]
  12. Dieleman, J.P.; Kerklaan, J.; Huygen, F.J.; Bouma, P.A.; Sturkenboom, M.C. Incidence rates and treatment of neuropathic pain conditions in the general population. Pain 2008, 137, 681–688. [Google Scholar] [CrossRef] [PubMed]
  13. Bouhassira, D.; Lanteri-Minet, M.; Attal, N.; Laurent, B.; Touboul, C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain 2008, 136, 380–387. [Google Scholar] [CrossRef] [PubMed]
  14. Attal, N.; Bouhassira, D. Pharmacotherapy of neuropathic pain: Which drugs, which treatment algorithms? Pain 2015, 156, S104–S114. [Google Scholar] [CrossRef] [PubMed]
  15. Dworkin, R.H.; Panarites, C.J.; Armstrong, E.P.; Malone, D.C.; Pham, S.V. Is treatment of postherpetic neuralgia in the community consistent with evidence-based recommendations? Pain 2012, 153, 869–875. [Google Scholar] [CrossRef] [PubMed]
  16. Torrance, N.; Ferguson, J.A.; Afolabi, E.; Bennett, M.I.; Serpell, M.G.; Dunn, K.M.; Smith, B.H. Neuropathic pain in the community: More under-treated than refractory? Pain 2013, 154, 690–699. [Google Scholar] [CrossRef] [PubMed]
  17. Calvo, M.; Dawes, J.M.; Bennett, D.L. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 2012, 11, 629–642. [Google Scholar] [CrossRef]
  18. Campbell, J.N.; Meyer, R.A. Mechanisms of neuropathic pain. Neuron 2006, 52, 77–92. [Google Scholar] [CrossRef] [PubMed]
  19. Sah, D.W.; Ossipo, M.H.; Porreca, F. Neurotrophic factors as novel therapeutics for neuropathic pain. Nat. Rev. Drug Discov. 2003, 2, 460–472. [Google Scholar] [CrossRef] [PubMed]
  20. 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]
  21. Ji, R.R.; Chamessian, A.; Zhang, Y.Q. Pain regulation by non-neuronal cells and inflammation. Science 2016, 354, 572–577. [Google Scholar] [CrossRef] [PubMed]
  22. Ren, K.; Dubner, R. Interactions between the immune and nervous systems in pain. Nat. Med. 2010, 16, 1267–1276. [Google Scholar] [CrossRef] [PubMed]
  23. Kiguchi, N.; Maeda, T.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. Macrophage inflammatory protein-1α mediates the development of neuropathic pain following peripheral nerve injury through interleukin-1β up-regulation. Pain 2010, 149, 305–315. [Google Scholar] [CrossRef] [PubMed]
  24. Mueller, M.; Wacker, K.; Ringelstein, E.B.; Hickey, W.F.; Imai, Y.; Kiefer, R. Rapid response of identified resident endoneurial macrophages to nerve injury. Am. J. Pathol. 2001, 159, 2187–2197. [Google Scholar] [CrossRef]
  25. Thacker, M.A.; Clark, A.K.; Marchand, F.; McMahon, S.B. Pathophysiology of peripheral neuropathic pain: Immune cells and molecules. Anesth. Analg. 2007, 105, 838–847. [Google Scholar] [CrossRef] [PubMed]
  26. Tofaris, G.K.; Patterson, P.H.; Jessen, K.R.; Mirsky, R. Denervated Schwann cells attract macrophages by secretion of leukemia inhibitory factor (LIF) and monocyte chemoattractant protein-1 in a process regulated by interleukin-6 and LIF. J. Neurosci. 2002, 22, 6696–6703. [Google Scholar] [PubMed]
  27. Zhang, F.F.; Morioka, N.; Harano, S.; Nakamura, Y.; Liu, K.; Nishibori, M.; Hisaoka-Nakashima, K.; Nakata, Y. Perineural expression of high-mobility group box-1 contributes to long-lasting mechanical hypersensitivity via matrix metalloproteinase-9 upregulation in mice with painful peripheral neuropathy. J. Neurochem. 2015, 136, 837–850. [Google Scholar] [CrossRef] [PubMed]
  28. Maeda, T.; Ozaki, M.; Kobayashi, Y.; Kiguchi, N.; Kishioka, S. HMGB1 as a potential therapeutic target for neuropathic pain. J. Pharmacol. Sci. 2013, 123, 301–305. [Google Scholar] [CrossRef] [PubMed]
  29. Scholz, J.; Woolf, C.J. The neuropathic pain triad: Neurons, immune cells and glia. Nat. Neurosci. 2007, 10, 1361–1368. [Google Scholar] [CrossRef] [PubMed]
  30. 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] [PubMed]
  31. Nicol, G.D.; Lopshire, J.C.; Pafford, C.M. Tumor necrosis factor enhances the capsaicin sensitivity of rat sensory neurons. J. Neurosci. 1997, 17, 975–982. [Google Scholar] [PubMed]
  32. Obreja, O.; Rathee, P.K.; Lips, K.S.; Distler, C.; Kress, M. IL-1 β potentiates heat-activated currents in rat sensory neurons: Involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J. 2002, 16, 1497–1503. [Google Scholar] [CrossRef] [PubMed]
  33. Oh, S.B.; Tran, P.B.; Gillard, S.E.; Hurley, R.W.; Hammond, D.L.; Miller, R.J. Chemokines and glycoprotein120 produce pain hypersensitivity by directly exciting primary nociceptive neurons. J. Neurosci. 2001, 21, 5027–5035. [Google Scholar] [PubMed]
  34. Zhang, N.; Inan, S.; Cowan, A.; Sun, R.; Wang, J.M.; Rogers, T.J.; Caterina, M.; Oppenheim, J.J. A proinflammatory chemokine, CCL3, sensitizes the heat- and capsaicin-gated ion channel TRPV1. Proc. Natl. Acad. Sci. USA 2005, 102, 4536–4541. [Google Scholar] [CrossRef] [PubMed]
  35. Kiguchi, N.; Kobayashi, Y.; Maeda, T.; Fukazawa, Y.; Tohya, K.; Kimura, M.; Kishioka, S. Epigenetic augmentation of the macrophage inflammatory protein 2/C-X-C chemokine receptor type 2 axis through histone H3 acetylation in injured peripheral nerves elicits neuropathic pain. J. Pharmacol. Exp. Ther. 2012, 340, 577–587. [Google Scholar] [CrossRef] [PubMed]
  36. Kobayashi, Y.; Kiguchi, N.; Fukazawa, Y.; Saika, F.; Maeda, T.; Kishioka, S. Macrophage-T cell interactions mediate neuropathic pain through the glucocorticoid-induced tumor necrosis factor ligand system. J. Biol. Chem. 2015, 290, 12603–12613. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, T.; van Rooijen, N.; Tracey, D.J. Depletion of macrophages reduces axonal degeneration and hyperalgesia following nerve injury. Pain 2000, 86, 25–32. [Google Scholar] [CrossRef]
  38. Ristoiu, V. Contribution of macrophages to peripheral neuropathic pain pathogenesis. Life Sci. 2013, 93, 870–881. [Google Scholar] [CrossRef] [PubMed]
  39. Basbaum, A.I.; Bautista, D.M.; Scherrer, G.; Julius, D. Cellular and molecular mechanisms of pain. Cell 2009, 139, 267–284. [Google Scholar] [CrossRef] [PubMed]
  40. Todd, A.J. Neuronal circuitry for pain processing in the dorsal horn. Nat. Rev. Neurosci. 2010, 11, 823–836. [Google Scholar] [CrossRef] [PubMed]
  41. Julius, D.; Basbaum, A.I. Molecular mechanisms of nociception. Nature 2001, 413, 203–210. [Google Scholar] [CrossRef] [PubMed]
  42. Dubin, A.E.; Patapoutian, A. Nociceptors: The sensors of the pain pathway. J. Clin. Investig. 2010, 120, 3760–3772. [Google Scholar] [CrossRef] [PubMed]
  43. Chahine, M.; Ziane, R.; Vijayaragavan, K.; Okamura, Y. Regulation of Na v channels in sensory neurons. Trends Pharmacol. Sci. 2005, 26, 496–502. [Google Scholar] [CrossRef] [PubMed]
  44. Moran, M.M.; McAlexander, M.A.; Biro, T.; Szallasi, A. Transient receptor potential channels as therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 601–620. [Google Scholar] [CrossRef] [PubMed]
  45. Woolf, C.J.; Ma, Q. Nociceptors–noxious stimulus detectors. Neuron 2007, 55, 353–364. [Google Scholar] [CrossRef] [PubMed]
  46. Waxman, S.G.; Zamponi, G.W. Regulating excitability of peripheral afferents: Emerging ion channel targets. Nat. Neurosci. 2014, 17, 153–163. [Google Scholar] [CrossRef] [PubMed]
  47. Dib-Hajj, S.D.; Black, J.A.; Waxman, S.G. Voltage-gated sodium channels: Therapeutic targets for pain. Pain Med. 2009, 10, 1260–1269. [Google Scholar] [CrossRef] [PubMed]
  48. 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] [PubMed]
  49. Xanthos, D.N.; Sandkuhler, J. Neurogenic neuroinflammation: Inflammatory CNS reactions in response to neuronal activity. Nat. Rev. Neurosci. 2014, 15, 43–53. [Google Scholar] [CrossRef] [PubMed]
  50. Pinho-Ribeiro, F.A.; Verri, W.A., Jr.; Chiu, I.M. Nociceptor Sensory Neuron-Immune Interactions in Pain and Inflammation. Trends Immunol. 2017, 38, 5–19. [Google Scholar] [CrossRef] [PubMed]
  51. White, F.A.; Sun, J.; Waters, S.M.; Ma, C.; Ren, D.; Ripsch, M.; Steflik, J.; Cortright, D.N.; Lamotte, R.H.; Miller, R.J. Excitatory monocyte chemoattractant protein-1 signaling is up-regulated in sensory neurons after chronic compression of the dorsal root ganglion. Proc. Natl. Acad. Sci. USA 2005, 102, 14092–14097. [Google Scholar] [CrossRef] [PubMed]
  52. Keller, A.F.; Beggs, S.; Salter, M.W.; De Koninck, Y. Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol. Pain 2007, 3, 27. [Google Scholar] [CrossRef] [PubMed]
  53. Haroutounian, S.; Nikolajsen, L.; Bendtsen, T.F.; Finnerup, N.B.; Kristensen, A.D.; Hasselstrom, J.B.; Jensen, T.S. Primary afferent input critical for maintaining spontaneous pain in peripheral neuropathy. Pain 2014, 155, 1272–1279. [Google Scholar] [CrossRef] [PubMed]
  54. Beggs, S.; Trang, T.; Salter, M.W. P2X4R+ microglia drive neuropathic pain. Nat. Neurosci. 2012, 15, 1068–1073. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. 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] [PubMed]
  57. McMahon, S.B.; Cafferty, W.B.; Marchand, F. Immune and glial cell factors as pain mediators and modulators. Exp. Neurol. 2005, 192, 444–462. [Google Scholar] [CrossRef] [PubMed]
  58. Tsuda, M.; Inoue, K.; Salter, M.W. Neuropathic pain and spinal microglia: A big problem from molecules in “small” glia. Trends Neurosci. 2005, 28, 101–107. [Google Scholar] [CrossRef] [PubMed]
  59. Ma, W.; Quirion, R. Partial sciatic nerve ligation induces increase in the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in astrocytes in the lumbar spinal dorsal horn and the gracile nucleus. Pain 2002, 99, 175–184. [Google Scholar] [CrossRef]
  60. Jin, S.X.; Zhuang, Z.Y.; Woolf, C.J.; Ji, R.R. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J. Neurosci. 2003, 23, 4017–4022. [Google Scholar] [PubMed]
  61. Tsuda, M.; Inoue, K. Neuron-microglia interaction by purinergic signaling in neuropathic pain following neurodegeneration. Neuropharmacology 2016, 104, 76–81. [Google Scholar] [CrossRef] [PubMed]
  62. Arruda, J.L.; Sweitzer, S.; Rutkowski, M.D.; DeLeo, J.A. Intrathecal anti-IL-6 antibody and IgG attenuates peripheral nerve injury-induced mechanical allodynia in the rat: Possible immune modulation in neuropathic pain. Brain Res. 2000, 879, 216–225. [Google Scholar] [CrossRef]
  63. Sweitzer, S.; Martin, D.; DeLeo, J.A. Intrathecal interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibits an anti-allodynic action in a rat model of neuropathic pain. Neuroscience 2001, 103, 529–539. [Google Scholar] [CrossRef]
  64. Thacker, M.A.; Clark, A.K.; Bishop, T.; Grist, J.; Yip, P.K.; Moon, L.D.; Thompson, S.W.; Marchand, F.; McMahon, S.B. CCL2 is a key mediator of microglia activation in neuropathic pain states. Eur. J. Pain 2009, 13, 263–272. [Google Scholar] [CrossRef] [PubMed]
  65. Kiguchi, N.; Kobayashi, Y.; Maeda, T.; Saika, F.; Kishioka, S. CC-chemokine MIP-1α in the spinal cord contributes to nerve injury-induced neuropathic pain. Neurosci. Lett. 2010, 484, 17–21. [Google Scholar] [CrossRef] [PubMed]
  66. Kuner, R. Central mechanisms of pathological pain. Nat. Med. 2010, 16, 1258–1266. [Google Scholar] [CrossRef] [PubMed]
  67. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [CrossRef] [PubMed]
  68. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef] [PubMed]
  69. Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef] [PubMed]
  70. Davies, L.C.; Jenkins, S.J.; Allen, J.E.; Taylor, P.R. Tissue-resident macrophages. Nat. Immunol. 2013, 14, 986–995. [Google Scholar] [CrossRef] [PubMed]
  71. Hussell, T.; Bell, T.J. Alveolar macrophages: Plasticity in a tissue-specific context. Nat. Rev. Immunol. 2014, 14, 81–93. [Google Scholar] [CrossRef] [PubMed]
  72. Pollard, J.W. Trophic macrophages in development and disease. Nat. Rev. Immunol. 2009, 9, 259–270. [Google Scholar] [CrossRef] [PubMed]
  73. Krenkel, O.; Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 2017, 17, 306–321. [Google Scholar] [CrossRef] [PubMed]
  74. Soehnlein, O.; Lindbom, L. Phagocyte partnership during the onset and resolution of inflammation. Nat. Rev. Immunol. 2010, 10, 427–439. [Google Scholar] [CrossRef] [PubMed]
  75. Wynn, T.A.; Vannella, K.M. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity 2016, 44, 450–462. [Google Scholar] [CrossRef] [PubMed]
  76. Ellis, A.; Bennett, D.L. Neuroinflammation and the generation of neuropathic pain. Br. J. Anaesth. 2013, 111, 26–37. [Google Scholar] [CrossRef] [PubMed]
  77. Perkins, N.M.; Tracey, D.J. Hyperalgesia due to nerve injury: Role of neutrophils. Neuroscience 2000, 101, 745–757. [Google Scholar] [CrossRef]
  78. Hu, P.; McLachlan, E.M. Macrophage and lymphocyte invasion of dorsal root ganglia after peripheral nerve lesions in the rat. Neuroscience 2002, 112, 23–38. [Google Scholar] [CrossRef]
  79. Moalem, G.; Xu, K.; Yu, L. T lymphocytes play a role in neuropathic pain following peripheral nerve injury in rats. Neuroscience 2004, 129, 767–777. [Google Scholar] [CrossRef] [PubMed]
  80. Morin, N.; Owolabi, S.A.; Harty, M.W.; Papa, E.F.; Tracy, T.F., Jr.; Shaw, S.K.; Kim, M.; Saab, C.Y. Neutrophils invade lumbar dorsal root ganglia after chronic constriction injury of the sciatic nerve. J. Neuroimmunol. 2007, 184, 164–171. [Google Scholar] [CrossRef] [PubMed]
  81. Kleinschnitz, C.; Hofstetter, H.H.; Meuth, S.G.; Braeuninger, S.; Sommer, C.; Stoll, G. T cell infiltration after chronic constriction injury of mouse sciatic nerve is associated with interleukin-17 expression. Exp. Neurol. 2006, 200, 480–485. [Google Scholar] [CrossRef] [PubMed]
  82. Peng, J.; Gu, N.; Zhou, L.; U, B.E.; Murugan, M.; Gan, W.B.; Wu, L.J. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nat. Commun. 2016, 7, 12029. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, W.; Quirion, R. Up-regulation of interleukin-6 induced by prostaglandin E from invading macrophages following nerve injury: An in vivo and in vitro study. J. Neurochem. 2005, 93, 664–673. [Google Scholar] [CrossRef] [PubMed]
  84. Gomez-Nicola, D.; Valle-Argos, B.; Suardiaz, M.; Taylor, J.S.; Nieto-Sampedro, M. Role of IL-15 in spinal cord and sciatic nerve after chronic constriction injury: Regulation of macrophage and T-cell infiltration. J. Neurochem. 2008, 107, 1741–1752. [Google Scholar] [CrossRef] [PubMed]
  85. Maeda, T.; Kiguchi, N.; Kobayashi, Y.; Ikuta, T.; Ozaki, M.; Kishioka, S. Leptin derived from adipocytes in injured peripheral nerves facilitates development of neuropathic pain via macrophage stimulation. Proc. Natl. Acad. Sci. USA 2009, 106, 13076–13081. [Google Scholar] [CrossRef] [PubMed]
  86. Conti, G.; Scarpini, E.; Baron, P.; Livraghi, S.; Tiriticco, M.; Bianchi, R.; Vedeler, C.; Scarlato, G. Macrophage infiltration and death in the nerve during the early phases of experimental diabetic neuropathy: A process concomitant with endoneurial induction of IL-1β and p75NTR. J. Neurol. Sci. 2002, 195, 35–40. [Google Scholar] [CrossRef]
  87. Yamagishi, S.; Ogasawara, S.; Mizukami, H.; Yajima, N.; Wada, R.; Sugawara, A.; Yagihashi, S. Correction of protein kinase C activity and macrophage migration in peripheral nerve by pioglitazone, peroxisome proliferator activated-γ-ligand, in insulin-deficient diabetic rats. J. Neurochem. 2008, 104, 491–499. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, C.C.; Lu, N.; Cui, Y.; Yang, T.; Zhao, Z.Q.; Xin, W.J.; Liu, X.G. Prevention of paclitaxel-induced allodynia by minocycline: Effect on loss of peripheral nerve fibers and infiltration of macrophages in rats. Mol. Pain 2010, 6, 76. [Google Scholar] [CrossRef] [PubMed]
  89. Kiguchi, N.; Maeda, T.; Kobayashi, Y.; Kondo, T.; Ozaki, M.; Kishioka, S. The critical role of invading peripheral macrophage-derived interleukin-6 in vincristine-induced mechanical allodynia in mice. Eur. J. Pharmacol. 2008, 592, 87–92. [Google Scholar] [CrossRef] [PubMed]
  90. Kiguchi, N.; Kobayashi, Y.; Kadowaki, Y.; Fukazawa, Y.; Saika, F.; Kishioka, S. Vascular endothelial growth factor signaling in injured nerves underlies peripheral sensitization in neuropathic pain. J. Neurochem. 2014, 129, 169–178. [Google Scholar] [CrossRef] [PubMed]
  91. Echeverry, S.; Wu, Y.; Zhang, J. Selectively reducing cytokine/chemokine expressing macrophages in injured nerves impairs the development of neuropathic pain. Exp. Neurol. 2013, 240, 205–218. [Google Scholar] [CrossRef] [PubMed]
  92. Ricardo, S.D.; van Goor, H.; Eddy, A.A. Macrophage diversity in renal injury and repair. J. Clin. Investig. 2008, 118, 3522–3530. [Google Scholar] [CrossRef] [PubMed]
  93. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef] [PubMed]
  94. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef] [PubMed]
  95. Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [PubMed]
  96. Krausgruber, T.; Blazek, K.; Smallie, T.; Alzabin, S.; Lockstone, H.; Sahgal, N.; Hussell, T.; Feldmann, M.; Udalova, I.A. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat. Immunol. 2011, 12, 231–238. [Google Scholar] [CrossRef] [PubMed]
  97. Lawrence, T.; Natoli, G. Transcriptional regulation of macrophage polarization: Enabling diversity with identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef] [PubMed]
  98. Satoh, T.; Takeuchi, O.; Vandenbon, A.; Yasuda, K.; Tanaka, Y.; Kumagai, Y.; Miyake, T.; Matsushita, K.; Okazaki, T.; Saitoh, T.; et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat. Immunol. 2010, 11, 936–944. [Google Scholar] [CrossRef] [PubMed]
  99. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef] [PubMed]
  100. Franco, R.; Fernandez-Suarez, D. Alternatively activated microglia and macrophages in the central nervous system. Prog. Neurobiol. 2015, 131, 65–86. [Google Scholar] [CrossRef] [PubMed]
  101. Benoit, M.; Desnues, B.; Mege, J.L. Macrophage polarization in bacterial infections. J. Immunol. 2008, 181, 3733–3739. [Google Scholar] [CrossRef] [PubMed]
  102. Hesketh, M.; Sahin, K.B.; West, Z.E.; Murray, R.Z. Macrophage Phenotypes Regulate Scar Formation and Chronic Wound Healing. Int. J. Mol. Sci. 2017, 18. [Google Scholar] [CrossRef] [PubMed]
  103. Kiguchi, N.; Kobayashi, Y.; Saika, F.; Sakaguchi, H.; Maeda, T.; Kishioka, S. Peripheral interleukin-4 ameliorates inflammatory macrophage-dependent neuropathic pain. Pain 2015, 156, 684–693. [Google Scholar] [CrossRef] [PubMed]
  104. Kiguchi, N.; Sakaguchi, H.; Kadowaki, Y.; Saika, F.; Fukazawa, Y.; Matsuzaki, S.; Kishioka, S. Peripheral administration of interleukin-13 reverses inflammatory macrophage and tactile allodynia in mice with partial sciatic nerve ligation. J. Pharmacol. Sci. 2017, 133, 53–56. [Google Scholar] [CrossRef] [PubMed]
  105. Komori, T.; Morikawa, Y.; Inada, T.; Hisaoka, T.; Senba, E. Site-specific subtypes of macrophages recruited after peripheral nerve injury. Neuroreport 2011, 22, 911–917. [Google Scholar] [CrossRef] [PubMed]
  106. Nathan, C.; Ding, A. Nonresolving inflammation. Cell 2010, 140, 871–882. [Google Scholar] [CrossRef] [PubMed]
  107. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [PubMed]
  108. Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef] [PubMed]
  109. Gouwy, M.; Struyf, S.; Proost, P.; Van Damme, J. Synergy in cytokine and chemokine networks amplifies the inflammatory response. Cytokine Growth Factor Rev. 2005, 16, 561–580. [Google Scholar] [CrossRef] [PubMed]
  110. Blanchet, X.; Langer, M.; Weber, C.; Koenen, R.R.; von Hundelshausen, P. Touch of chemokines. Front. Immunol. 2012, 3, 175. [Google Scholar] [CrossRef] [PubMed]
  111. Liu, X.; Fang, L.; Guo, T.B.; Mei, H.; Zhang, J.Z. Drug targets in the cytokine universe for autoimmune disease. Trends Immunol. 2013, 34, 120–128. [Google Scholar] [CrossRef] [PubMed]
  112. Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59. [Google Scholar] [CrossRef] [PubMed]
  113. Zelenka, M.; Schafers, M.; Sommer, C. Intraneural injection of interleukin-1beta and tumor necrosis factor-alpha into rat sciatic nerve at physiological doses induces signs of neuropathic pain. Pain 2005, 116, 257–263. [Google Scholar] [CrossRef] [PubMed]
  114. Wei, X.H.; Zang, Y.; Wu, C.Y.; Xu, J.T.; Xin, W.J.; Liu, X.G. Peri-sciatic administration of recombinant rat TNF-α induces mechanical allodynia via upregulation of TNF-α in dorsal root ganglia and in spinal dorsal horn: The role of NF-κ B pathway. Exp. Neurol. 2007, 205, 471–484. [Google Scholar] [CrossRef] [PubMed]
  115. Shamash, S.; Reichert, F.; Rotshenker, S. The cytokine network of Wallerian degeneration: Tumor necrosis factor-α, interleukin-1α, and interleukin-1β. J. Neurosci. 2002, 22, 3052–3060. [Google Scholar] [PubMed]
  116. Ramer, M.S.; Murphy, P.G.; Richardson, P.M.; Bisby, M.A. Spinal nerve lesion-induced mechanoallodynia and adrenergic sprouting in sensory ganglia are attenuated in interleukin-6 knockout mice. Pain 1998, 78, 115–121. [Google Scholar] [CrossRef]
  117. Schafers, M.; Svensson, C.I.; Sommer, C.; Sorkin, L.S. Tumor necrosis factor-α induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons. J. Neurosci. 2003, 23, 2517–2521. [Google Scholar] [PubMed]
  118. Wolf, G.; Gabay, E.; Tal, M.; Yirmiya, R.; Shavit, Y. Genetic impairment of interleukin-1 signaling attenuates neuropathic pain, autotomy, and spontaneous ectopic neuronal activity, following nerve injury in mice. Pain 2006, 120, 315–324. [Google Scholar] [CrossRef] [PubMed]
  119. Palmqvist, C.; Wardlaw, A.J.; Bradding, P. Chemokines and their receptors as potential targets for the treatment of asthma. Br. J. Pharmacol. 2007, 151, 725–736. [Google Scholar] [CrossRef] [PubMed]
  120. Ransohoff, R.M. Chemokines and chemokine receptors: Standing at the crossroads of immunobiology and neurobiology. Immunity 2009, 31, 711–721. [Google Scholar] [CrossRef] [PubMed]
  121. Biber, K.; Boddeke, E. Neuronal CC chemokines: The distinct roles of CCL21 and CCL2 in neuropathic pain. Front. Cell. Neurosci. 2014, 8, 210. [Google Scholar] [CrossRef] [PubMed]
  122. Dawes, J.M.; McMahon, S.B. Chemokines as peripheral pain mediators. Neurosci. Lett. 2013, 557 Pt A, 1–8. [Google Scholar] [CrossRef] [PubMed]
  123. Kiguchi, N.; Saika, F.; Kobayashi, Y.; Kishioka, S. Epigenetic regulation of CC-chemokine ligand 2 in nonresolving inflammation. Biomol. Concepts 2014, 5, 265–273. [Google Scholar] [CrossRef] [PubMed]
  124. Taskinen, H.S.; Roytta, M. Increased expression of chemokines (MCP-1, MIP-1α, RANTES) after peripheral nerve transection. J. Peripher. Nerv. Syst. 2000, 5, 75–81. [Google Scholar] [CrossRef] [PubMed]
  125. Kiguchi, N.; Kobayashi, Y.; Saika, F.; Kishioka, S. Epigenetic upregulation of CCL2 and CCL3 via histone modifications in infiltrating macrophages after peripheral nerve injury. Cytokine 2013, 64, 666–672. [Google Scholar] [CrossRef] [PubMed]
  126. Abbadie, C.; Lindia, J.A.; Cumiskey, A.M.; Peterson, L.B.; Mudgett, J.S.; Bayne, E.K.; DeMartino, J.A.; MacIntyre, D.E.; Forrest, M.J. Impaired neuropathic pain responses in mice lacking the chemokine receptor CCR2. Proc. Natl. Acad. Sci. USA 2003, 100, 7947–7952. [Google Scholar] [CrossRef] [PubMed]
  127. Padi, S.S.; 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] [PubMed]
  128. White, F.A.; Feldman, P.; Miller, R.J. Chemokine signaling and the management of neuropathic pain. Mol. Interv. 2009, 9, 188–195. [Google Scholar] [CrossRef] [PubMed]
  129. Saika, F.; Kiguchi, N.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. CC-chemokine ligand 4/macrophage inflammatory protein-1beta participates in the induction of neuropathic pain after peripheral nerve injury. Eur. J. Pain 2012, 16, 1271–1280. [Google Scholar] [CrossRef] [PubMed]
  130. Liou, J.T.; Yuan, H.B.; Mao, C.C.; Lai, Y.S.; Day, Y.J. Absence of C-C motif chemokine ligand 5 in mice leads to decreased local macrophage recruitment and behavioral hypersensitivity in a murine neuropathic pain model. Pain 2012, 153, 1283–1291. [Google Scholar] [CrossRef] [PubMed]
  131. Sun, J.H.; Yang, B.; Donnelly, D.F.; Ma, C.; LaMotte, R.H. MCP-1 enhances excitability of nociceptive neurons in chronically compressed dorsal root ganglia. J. Neurophysiol. 2006, 96, 2189–2199. [Google Scholar] [CrossRef] [PubMed]
  132. Fenini, G.; Contassot, E.; French, L.E. Potential of IL-1, IL-18 and Inflammasome Inhibition for the Treatment of Inflammatory Skin Diseases. Front. Pharmacol. 2017, 8, 278. [Google Scholar] [CrossRef] [PubMed]
  133. Croft, M.; Siegel, R.M. Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat. Rev. Rheumatol. 2017, 13, 217–233. [Google Scholar] [CrossRef] [PubMed]
  134. Martin-Blondel, G.; Brassat, D.; Bauer, J.; Lassmann, H.; Liblau, R.S. CCR5 blockade for neuroinflammatory diseases—Beyond control of HIV. Nat. Rev. Neurol. 2016, 12, 95–105. [Google Scholar] [CrossRef] [PubMed]
  135. Dinarello, C.A.; Joosten, L.A. Inflammation in rheumatology in 2015: New tools to tackle inflammatory arthritis. Nat. Rev. Rheumatol. 2016, 12, 78–80. [Google Scholar] [CrossRef] [PubMed]
  136. Chatila, T.A. Interleukin-4 receptor signaling pathways in asthma pathogenesis. Trends Mol. Med. 2004, 10, 493–499. [Google Scholar] [CrossRef] [PubMed]
  137. Gandhi, N.A.; Bennett, B.L.; Graham, N.M.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2 inflammation in disease. Nat. Rev. Drug Discov. 2016, 15, 35–50. [Google Scholar] [CrossRef] [PubMed]
  138. Gause, W.C.; Wynn, T.A.; Allen, J.E. Type 2 immunity and wound healing: Evolutionary refinement of adaptive immunity by helminths. Nat. Rev. Immunol. 2013, 13, 607–614. [Google Scholar] [CrossRef] [PubMed]
  139. Wynn, T.A. Type 2 cytokines: Mechanisms and therapeutic strategies. Nat. Rev. Immunol. 2015, 15, 271–282. [Google Scholar] [CrossRef] [PubMed]
  140. West, G.A.; Matsuura, T.; Levine, A.D.; Klein, J.S.; Fiocchi, C. Interleukin 4 in inflammatory bowel disease and mucosal immune reactivity. Gastroenterology 1996, 110, 1683–1695. [Google Scholar] [CrossRef] [PubMed]
  141. Shaw, M.K.; Lorens, J.B.; Dhawan, A.; DalCanto, R.; Tse, H.Y.; Tran, A.B.; Bonpane, C.; Eswaran, S.L.; Brocke, S.; Sarvetnick, N.; et al. Local delivery of interleukin 4 by retrovirus-transduced T lymphocytes ameliorates experimental autoimmune encephalomyelitis. J. Exp. Med. 1997, 185, 1711–1714. [Google Scholar] [CrossRef] [PubMed]
  142. Lubberts, E.; Joosten, L.A.; Chabaud, M.; van Den Bersselaar, L.; Oppers, B.; Coenen-De Roo, C.J.; Richards, C.D.; Miossec, P.; van Den Berg, W.B. IL-4 gene therapy for collagen arthritis suppresses synovial IL-17 and osteoprotegerin ligand and prevents bone erosion. J. Clin. Investig. 2000, 105, 1697–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Liou, J.T.; Lee, C.M.; Lin, Y.C.; Chen, C.Y.; Liao, C.C.; Lee, H.C.; Day, Y.J. P-selectin is required for neutrophils and macrophage infiltration into injured site and contributes to generation of behavioral hypersensitivity following peripheral nerve injury in mice. Pain 2013, 154, 2150–2159. [Google Scholar] [CrossRef] [PubMed]
  144. Grace, P.M.; Fabisiak, T.J.; Green-Fulgham, S.M.; Anderson, N.D.; Strand, K.A.; Kwilasz, A.J.; Galer, E.L.; Walker, F.R.; Greenwood, B.N.; Maier, S.F.; et al. Prior voluntary wheel running attenuates neuropathic pain. Pain 2016, 157, 2012–2023. [Google Scholar] [CrossRef] [PubMed]
  145. Saika, F.; Kiguchi, N.; Kobayashi, Y.; Kishioka, S. Peripheral α4β2 nicotinic acetylcholine receptor signalling attenuates tactile allodynia and thermal hyperalgesia after nerve injury in mice. Acta Physiol. 2015, 213, 462–471. [Google Scholar] [CrossRef] [PubMed]
  146. Ulloa, L. The vagus nerve and the nicotinic anti-inflammatory pathway. Nat. Rev. Drug Discov. 2005, 4, 673–684. [Google Scholar] [CrossRef] [PubMed]
  147. Dinarello, C.A. The interleukin-1 family: 10 years of discovery. FASEB J. 1994, 8, 1314–1325. [Google Scholar] [PubMed]
  148. Van Assche, G.; Rutgeerts, P. Anti-TNF agents in Crohn’s disease. Expert Opin. Investig. Drugs 2000, 9, 103–111. [Google Scholar] [CrossRef] [PubMed]
  149. Feldmann, M. Development of anti-TNF therapy for rheumatoid arthritis. Nat. Rev. Immunol. 2002, 2, 364–371. [Google Scholar] [CrossRef] [PubMed]
  150. Okoro, T.; Tafazal, S.I.; Longworth, S.; Sell, P.J. Tumor necrosis α-blocking agent (etanercept): A triple blind randomized controlled trial of its use in treatment of sciatica. J. Spinal Disord. Tech. 2010, 23, 74–77. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, Y.F.; Chen, P.Y.; Chang, W.; Zhu, F.Q.; Xu, L.L.; Wang, S.L.; Chang, L.Y.; Luo, J.; Liu, G.J. Clinical significance of tumor necrosis factor-alpha inhibitors in the treatment of sciatica: A systematic review and meta-analysis. PLoS ONE 2014, 9, e103147. [Google Scholar]
  152. Fisher, C.J., Jr.; Dhainaut, J.F.; Opal, S.M.; Pribble, J.P.; Balk, R.A.; Slotman, G.J.; Iberti, T.J.; Rackow, E.C.; Shapiro, M.J.; Greenman, R.L.; et al. Recombinant human interleukin 1 receptor antagonist in the treatment of patients with sepsis syndrome. Results from a randomized, double-blind, placebo-controlled trial. Phase III rhIL-1ra Sepsis Syndrome Study Group. JAMA 1994, 271, 1836–1843. [Google Scholar] [CrossRef] [PubMed]
  153. Abraham, E.; Anzueto, A.; Gutierrez, G.; Tessler, S.; San Pedro, G.; Wunderink, R.; Dal Nogare, A.; Nasraway, S.; Berman, S.; Cooney, R.; et al. Double-blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. NORASEPT II Study Group. Lancet 1998, 351, 929–933. [Google Scholar] [CrossRef]
  154. Pavlov, V.A.; Tracey, K.J. Neural regulation of immunity: Molecular mechanisms and clinical translation. Nat. Neurosci. 2017, 20, 156–166. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, H.; Yu, M.; Ochani, M.; Amella, C.A.; Tanovic, M.; Susarla, S.; Li, J.H.; Wang, H.; Yang, H.; Ulloa, L.; et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation. Nature 2003, 421, 384–388. [Google Scholar] [CrossRef] [PubMed]
  156. De Jonge, W.J.; van der Zanden, E.P.; The, F.O.; Bijlsma, M.F.; van Westerloo, D.J.; Bennink, R.J.; Berthoud, H.R.; Uematsu, S.; Akira, S.; van den Wijngaard, R.M.; et al. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat. Immunol. 2005, 6, 844–851. [Google Scholar] [CrossRef] [PubMed]
  157. Tracey, K.J. Physiology and immunology of the cholinergic antiinflammatory pathway. J. Clin. Investig. 2007, 117, 289–296. [Google Scholar] [CrossRef] [PubMed]
  158. Borovikova, L.V.; Ivanova, S.; Zhang, M.; Yang, H.; Botchkina, G.I.; Watkins, L.R.; Wang, H.; Abumrad, N.; Eaton, J.W.; Tracey, K.J. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000, 405, 458–462. [Google Scholar] [PubMed]
  159. Wang, H.; Liao, H.; Ochani, M.; Justiniani, M.; Lin, X.; Yang, L.; Al-Abed, Y.; Wang, H.; Metz, C.; Miller, E.J.; et al. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat. Med. 2004, 10, 1216–1221. [Google Scholar] [CrossRef] [PubMed]
  160. Saeed, R.W.; Varma, S.; Peng-Nemeroff, T.; Sherry, B.; Balakhaneh, D.; Huston, J.; Tracey, K.J.; Al-Abed, Y.; Metz, C.N. Cholinergic stimulation blocks endothelial cell activation and leukocyte recruitment during inflammation. J. Exp. Med. 2005, 201, 1113–1123. [Google Scholar] [CrossRef] [PubMed]
  161. Lindstrom, J. Nicotinic acetylcholine receptors in health and disease. Mol. Neurobiol. 1997, 15, 193–222. [Google Scholar] [CrossRef] [PubMed]
  162. Miwa, J.M.; Freedman, R.; Lester, H.A. Neural systems governed by nicotinic acetylcholine receptors: Emerging hypotheses. Neuron 2011, 70, 20–33. [Google Scholar] [CrossRef] [PubMed]
  163. Van der Zanden, E.P.; Snoek, S.A.; Heinsbroek, S.E.; Stanisor, O.I.; Verseijden, C.; Boeckxstaens, G.E.; Peppelenbosch, M.P.; Greaves, D.R.; Gordon, S.; De Jonge, W.J. Vagus nerve activity augments intestinal macrophage phagocytosis via nicotinic acetylcholine receptor α4β2. Gastroenterology 2009, 137, 1029–1039. [Google Scholar] [CrossRef] [PubMed]
  164. Nemethova, A.; Michel, K.; Gomez-Pinilla, P.J.; Boeckxstaens, G.E.; Schemann, M. Nicotine attenuates activation of tissue resident macrophages in the mouse stomach through the β2 nicotinic acetylcholine receptor. PLoS ONE 2013, 8, e79264. [Google Scholar] [CrossRef] [PubMed]
  165. Kiguchi, N.; Kobayashi, Y.; Maeda, T.; Tominaga, S.; Nakamura, J.; Fukazawa, Y.; Ozaki, M.; Kishioka, S. Activation of nicotinic acetylcholine receptors on bone marrow-derived cells relieves neuropathic pain accompanied by peripheral neuroinflammation. Neurochem. Int. 2012, 61, 1212–1219. [Google Scholar] [CrossRef] [PubMed]
  166. Kiguchi, N.; Saika, F.; Kobayashi, Y.; Ko, M.C.; Kishioka, S. TC-2559, an α4β2 nicotinic acetylcholine receptor agonist, suppresses the expression of CCL3 and IL-1β through STAT3 inhibition in cultured murine macrophages. J. Pharmacol. Sci. 2015, 128, 83–86. [Google Scholar] [CrossRef] [PubMed]
  167. Kiguchi, N.; Maeda, T.; Kobayashi, Y.; Fukazawa, Y.; Kishioka, S. Leptin enhances CC-chemokine ligand expression in cultured murine macrophage. Biochem. Biophys. Res. Commun. 2009, 384, 311–315. [Google Scholar] [CrossRef] [PubMed]
  168. Samavati, L.; Rastogi, R.; Du, W.; Huttemann, M.; Fite, A.; Franchi, L. STAT3 tyrosine phosphorylation is critical for interleukin 1 β and interleukin-6 production in response to lipopolysaccharide and live bacteria. Mol. Immunol. 2009, 46, 1867–1877. [Google Scholar] [CrossRef] [PubMed]
  169. Burke, J.G.; Watson, R.W.; McCormack, D.; Dowling, F.E.; Walsh, M.G.; Fitzpatrick, J.M. Spontaneous production of monocyte chemoattractant protein-1 and interleukin-8 by the human lumbar intervertebral disc. Spine 2002, 27, 1402–1407. [Google Scholar] [CrossRef] [PubMed]
  170. Purwata, T.E. High TNF-α plasma levels and macrophages iNOS and TNF-α expression as risk factors for painful diabetic neuropathy. J. Pain Res. 2011, 4, 169–175. [Google Scholar] [CrossRef] [PubMed]
  171. Lenz, M.; Uceyler, N.; Frettloh, J.; Hoffken, O.; Krumova, E.K.; Lissek, S.; Reinersmann, A.; Sommer, C.; Stude, P.; Waaga-Gasser, A.M.; et al. Local cytokine changes in complex regional pain syndrome type I (CRPS I) resolve after 6 months. Pain 2013, 154, 2142–2149. [Google Scholar] [CrossRef] [PubMed]
  172. Backryd, E.; Ghafouri, B.; Larsson, B.; Gerdle, B. Plasma pro-inflammatory markers in chronic neuropathic pain: A multivariate, comparative, cross-sectional pilot study. Scand. J. Pain 2016, 10, 1–5. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow cytometric analysis of leukocytes infiltrating injured nerves. Mice were subjected to partial sciatic nerve ligation, and the sciatic nerves were isolated on day 7 after injury. Representative forward scatter (FSC) versus side scatter (SSC) plots show total events collected from control (A) or injured (B) sciatic nerves. Representative plots of CD11b versus F4/80 from the gates of each FSC/SSC plot demonstrate that CD11b+ F4/80+ macrophages are much more abundant in the injured nerves (D) than in the control nerves (C). Representative plots of Ly6G versus CD3 from the F4/80 population indicate a similar increase in infiltrating Ly6G+ neutrophils and CD3+ T lymphocytes in the injured (F) compared with the control (E) nerves.
Figure 1. Flow cytometric analysis of leukocytes infiltrating injured nerves. Mice were subjected to partial sciatic nerve ligation, and the sciatic nerves were isolated on day 7 after injury. Representative forward scatter (FSC) versus side scatter (SSC) plots show total events collected from control (A) or injured (B) sciatic nerves. Representative plots of CD11b versus F4/80 from the gates of each FSC/SSC plot demonstrate that CD11b+ F4/80+ macrophages are much more abundant in the injured nerves (D) than in the control nerves (C). Representative plots of Ly6G versus CD3 from the F4/80 population indicate a similar increase in infiltrating Ly6G+ neutrophils and CD3+ T lymphocytes in the injured (F) compared with the control (E) nerves.
Ijms 18 02296 g001
Figure 2. Generation of neuropathic pain by macrophage-driven inflammation in the peripheral nervous system. After nerve injury, activated resident cells (Schwann cells and macrophages) produce soluble factors such as damage-associated molecular patterns (DAMPs) that activate nearby cells and recruit circulating leukocytes (macrophages, neutrophils, and lymphocytes) to the site of injury. Macrophages are the most abundant infiltrating leukocyte population and are thought to play a central role in regulating peripheral neuroinflammation. Tissue-resident and infiltrating leukocytes communicate through the release of pro-inflammatory mediators such as cytokines and chemokines, which convey nociceptive information to dorsal root ganglia (DRG) neurons. Persistent ectopic activity of DRG neurons induces central sensitization characterized by the enhanced activity of pain-processing neurons and the activation of microglia and astrocytes. Pharmacological targeting of macrophages or macrophage-derived pro-inflammatory molecules by nicotinic acetylcholine receptor (nAChR) ligands, Th2 cytokines, and inhibitors of cytokines and chemokines can suppress macrophage-driven neuroinflammation after nerve injury. The reduction in neuroinflammation improves both peripheral and central sensitization and alleviates intractable neuropathic pain.
Figure 2. Generation of neuropathic pain by macrophage-driven inflammation in the peripheral nervous system. After nerve injury, activated resident cells (Schwann cells and macrophages) produce soluble factors such as damage-associated molecular patterns (DAMPs) that activate nearby cells and recruit circulating leukocytes (macrophages, neutrophils, and lymphocytes) to the site of injury. Macrophages are the most abundant infiltrating leukocyte population and are thought to play a central role in regulating peripheral neuroinflammation. Tissue-resident and infiltrating leukocytes communicate through the release of pro-inflammatory mediators such as cytokines and chemokines, which convey nociceptive information to dorsal root ganglia (DRG) neurons. Persistent ectopic activity of DRG neurons induces central sensitization characterized by the enhanced activity of pain-processing neurons and the activation of microglia and astrocytes. Pharmacological targeting of macrophages or macrophage-derived pro-inflammatory molecules by nicotinic acetylcholine receptor (nAChR) ligands, Th2 cytokines, and inhibitors of cytokines and chemokines can suppress macrophage-driven neuroinflammation after nerve injury. The reduction in neuroinflammation improves both peripheral and central sensitization and alleviates intractable neuropathic pain.
Ijms 18 02296 g002

Share and Cite

MDPI and ACS Style

Kiguchi, N.; Kobayashi, D.; Saika, F.; Matsuzaki, S.; Kishioka, S. Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages. Int. J. Mol. Sci. 2017, 18, 2296. https://doi.org/10.3390/ijms18112296

AMA Style

Kiguchi N, Kobayashi D, Saika F, Matsuzaki S, Kishioka S. Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages. International Journal of Molecular Sciences. 2017; 18(11):2296. https://doi.org/10.3390/ijms18112296

Chicago/Turabian Style

Kiguchi, Norikazu, Daichi Kobayashi, Fumihiro Saika, Shinsuke Matsuzaki, and Shiroh Kishioka. 2017. "Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages" International Journal of Molecular Sciences 18, no. 11: 2296. https://doi.org/10.3390/ijms18112296

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