Abstract
Microglial cells are specialized macrophage cells of the central nervous system responsible for the innate immunity of the spinal cord and the brain. They protect the brain and spinal cord from invaders, microbes, demyelination, trauma and remove defective cells and neurons. For immune protection, microglial cells possess a significant number of receptors and chemical mediators that allow them to communicate rapidly and specifically with all cells of the nervous tissue. The contribution of microglia in neuropathic pain challenges conventional concepts toward neurons being the only structure responsible for the pathophysiological changes that drive neuropathic pain. The present study is a narrative review focusing on the literature concerning the complex interaction between neurons and microglia in the development of neuropathic pain. Injury in the peripheral or central nervous system may result in maladaptive changes in neurons and microglial cells. In neuropathic pain, microglial cells have an important role in initiating and maintenance of pain and inflammation. The interaction between neural and microglial cells has been proven extremely crucial for chronic pain. The study of individual mechanisms at the level of the spinal cord and the brain is an interesting and groundbreaking research challenge. Elucidation of the mechanisms by which neurons and immune cells interact, could constitute microglial cells a new therapeutic target for the treatment of neuropathic pain.
Keywords: spinal cord, neuroinflammation, neuropathic pain, microglial cells, microglia
Introduction and background
Neuropathic pain is defined by the International Association for the Study of Pain as “pain caused by a lesion or a disease of the somatosensory nervous system.” Unlike nociceptive pain, which is the normal response to noxious stimuli, neuropathic pain is a maladaptive response to an injury of the nervous system [1]. In neuropathic pain, which can be characterized as acute or chronic and central or peripheral, tissue damage is not always obvious. The most common causes of neuropathic pain include trigeminal neuralgia (idiopathic), post-amputation pain, persistent post-operative or post-traumatic pain, post-herpetic neuralgia after a herpes zoster infection, painful diabetic polyneuropathy, pain in patients with cancer and those infected by HIV, and pain as a side effect of certain medications (chemotherapy) [2]. As pain is a subjective measurement, diagnosis of neuropathic pain is difficult. Common features of neuropathic pain are the presence of allodynia, where a normally non-painful stimulus results in pain, and hyperalgesia, where there is an increased response to a painful stimulus, and spontaneous pain [3].
Regardless of etiology, location, intensity, or other individual characteristics, neuropathic pain is characterized by changes in the normal processing of sensory signals. Despite the absence of tissue damage, the pain is maintained through the development of peripheral and central maladaptive mechanisms. The disorder develops gradually for weeks or months after damage, decisively affecting patients’ quality of life. Once neuropathic pain is generated, the sensory hypersensitivity typically persists for prolonged periods, even though the original etiological cause may have long since disappeared, as after nerve trauma. Symptom persistence ultimately causes changes in the genetic expression of the neurons of the somatosensory system and extension of the cortical representation in areas of the primary sensory cortex [1]. First-line treatment modalities for neuropathic pain do not target the underlying mechanisms causing the pain and are not specific to the different phases in the development of neuropathic pain [4,5].
Microglia, first described in 1920 by del Río Hortega, are a specialized population of mononuclear macrophages that are located in the central nervous system (CNS) and comprise ~10-15% of all glial cells. They participate in immune surveillance and homeostatic control of inflammation and regulate the innate immune response of the brain and spinal cord. They develop from the progenitor hematopoietic stem cells (myeloid progenitor cells) during early embryogenesis and migrate initially to the brain in relatively small numbers [6]. However, their impressive proliferation ability allows them to rapidly colonize the entire CNS. This ability is maintained eternally through the colony-stimulating factor 1 (CSF-1), which is expressed in the membrane of microglial cells in the adult brain and is associated with the expansion of the microglia population during neuroinflammation and neuropathic pain [7].
Microglial cells are considered as the fastest and most flexible cells in the brain. They have a small cell body and several long processes with multiple branches that allow them to monitor each neuron individually while "slipping" among them by extending and shrinking their branches [8]. They are oriented in space and are attracted by ischemia or brain damage. They constantly control all spinal and cerebral regions, the synapses of each neuron, and each neural circuit [9]. They stay in touch with each of the synapses for a few minutes and then move on to the next one. They continuously evaluate the usefulness of the neural circuits, prioritizing frequently used synapses and completing the scanning of billions of cerebral and spinal synapses within a few hours. This constant vigilance of microglial cells protects the brain and spinal cord from invaders, germs, demyelination, trauma, and mutated or defective cells [10]. Microglia respond quickly with morphological changes when stimuli affect the physiological homeostasis of the CNS: microglia numbers increase, volume increases, processes retract and become ramified. Activated microglia have two polarization types: M1-type (M1) and M2-type (M2), pro-inflammatory and anti-inflammatory, respectively. M1 microglia have a strong phagocytic ability and can produce many pro-inflammatory factors [11].
The present study is a narrative review focusing on the literature concerning the complex interaction between neurons and microglia in the development of neuropathic pain.
Review
Following a peripheral nerve injury, the system of pain management and treatment malfunctions. Research efforts and pharmacological approaches have focused on neural tissue dysfunction. However, evidence suggests that in addition to nerve cells, specialized non-neural cells that regulate the innate immune response of neural tissue against pathogens, play an important role in this dysfunction, which ultimately leads to neuropathic pain [12-14]. An increase in the number of microglial cells in the spinal cord after peripheral nerve damage has been reported since 1970 [15], and perhaps this action of glial cells is the missing element to explain the conversion of acute pain to chronic [16,17].
Laboratory experiments using modern tools of molecular genetics have shown that microglia participate in the onset, maintenance, mediation, and expansion of neuropathic pain [13,18]. In models of neuropathic pain (compression of the peripheral or spinal nerve), inhibition of microglial cell activity led to milder pain. Advanced research techniques (transgenic and chimeric animal models of gene targeting) have shown that nerve damage activates microglia causing rapid changes in morphology, migration rate, and proliferation [19]. A recent study used chemogenetic approach as a novel tool to examine the role of microglia in neuropathic pain in mice. Researchers used transgenic mice and demonstrated that microglial Gi signaling by chemogenetic manipulation attenuates chronic pain via inhibition of neuroinflammation [20]. An increase in the number of protein molecules on the surface of microglia (more than 40 have been identified) after peripheral nerve damage, indicates that microglia is one of the contributors to neuropathic pain [11,21].
Neuroglial cells (microglia, astrocytes, oligodendrocytes, and ependymal cells), besides the support and nutrition of neurons, actively participate in the reception, transmission, processing, and modification of nerve stimuli. Glial cells and neurons closely cooperate aiming at homeostasis and optimal functioning of the sensory synapses. In pain and tissue damage, neurons are assisted by glial cells that are activated at the dorsal root ganglia and the posterior horns of the spinal cord. Glial cells are co-responsible to some extent for the expansion of pain. Many non-opioid and opioid peptides act on microglial cells, which respond by producing pro- or anti-inflammatory agents to a continuous supplementation of neurons with excitatory chemical signals [22,23].
From the peripheral nerves to the spinal cord
In nerve damage (trauma, neuropathy, surgery, etc.), inflammatory irritants fill the space around the free nerve endings, peripheral receptors, axons, or peripheral stumps. The sensory nerves, through their special channels, including transient receptor potential vanilloid 1 (TRPV1), prostaglandin receptors, tendon-sensitive ion channels, etc.), convert chemical stimuli into electrical energy potentials. Glial cells around the spinal synapses respond to both electrical and chemical stimuli; tensile cells in the cell membrane "sense" the intensity of nerve activity and respond to it with hyperstimulation. Astrocytes and microglial cells, which are scattered but regularly arranged throughout the CNS and along the somatosensory pathway, express receptors for most of the substances that act on the spinal sensory synapsis. On the membrane of glial cells, there are glutamic acid receptors (ionotropic: N-methyl-D-aspartate or NMDA and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid or AMPA; metabotropic: mGluR I, II, III), gamma-aminobutyric acid receptors (GABA A & GABA B), norepinephrine receptors (α1, α2, β1, and β2 inactivated protein), acetylcholine receptors (nicotinic α3, α5, α6, α7, and β4 receptors), cannabinoid receptors (CB2) and receptors of bradykinin, dopamine (D1 & D2), purines, adenosine, and opioids [24].
Some transmembrane receptors are expressed in resting microglia and others in the activated state. Toll-like receptor 4 is overexpressed in microglial cells after their activation. Its overexpression promotes the production of nitric oxides, prostaglandins, leukotrienes, nerve growth factors, stimulant amino acids, neurotoxic peroxides, and cytokines, such as interleukin-1 (IL-1), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). These substances further stimulate the perisynaptic region in the microglia-axons positive feedback loop. This toxic compound of chemical substances originating from nerve cells, astrocytes, and microglial cells is responsible for chronic and persistent neuroinflammation [25-27].
Evidence has led to the hypothesis of "microgliopathic pain," a pain syndrome for which dysfunctional microglia are responsible. It has been thought that the theory of dysfunctional microglia in chronic pain may be the "missing link" in pain physiology. Dysregulated microglial activation, neuroinflammation, and altered neuronal communication contribute to the amplification of pain signals and the transition from acute to chronic pain. By targeting microglial-related mechanisms, researchers are striving to develop effective treatments that can provide relief to individuals suffering from chronic pain [28].
Events in the spinal cord
After peripheral nerve damage, the resting microglia of the spinal cord rapidly changes both morphologically and functionally. Resting microglial cells have small cell bodies and long-branched processes. As microglial cells are activated, the cells undergo morphological changes in which the cell body becomes relatively large, and the processes become shorter. These changes are accompanied by an increase in the number of microglia in the spinal cord, either by migration or proliferation, a phenomenon known as gliosis or microgliosis [29]. These activated microglial cells, also called amoeboid microglia, have higher rates of migration, proliferation, and phagocytic behavior. Besides the morphological changes in microglial phenotype, an increase in surface immunogenic antigens and the production of cytotoxic and neurotrophic factors has been reported [30-32].
Besides cellular hypertrophy, overexpression of the complement receptor CR3 on the microglial cell surface is presented, changes that are observed within the first 24 hours of nerve damage. Two to three days after nerve injury, microglia begin to proliferate, reaching the maximum number of cells on the 4th to 7th day. Within the first seven days, the most prominent changes in the immunophenotype are observed, with more frequent increased expression of major histocompatibility complex (MHC) antigens. The changes are reversible regardless of the severity of the damage and usually are over one month after the start of the damage. Temporal continuity was one of the reasons for accepting the role of microglia in the structural changes of sensory cells and in the genesis of neuropathic pain [33-36].
CSF-1 has been identified as the key chemoattractant of microglial cells, which invites them to colonize and expand at the site of the lesion. Concurrently, its receptor, CSF1R, located on the surface of glial cells of the spinal cord, increases in number proportionally [37]. It simultaneously interacts with a) DNAX-activating protein 12 (DAP12), a transmembrane signal transduction protein responsible for microglial cell proliferation, and b) a second DAP12-dependent signaling pathway through which some specific microglial genes associated with pain sensitivity are up-regulated, such as interferon regulating factor 8 (IRF8) and 5 (IRF5). The expression of IRF8 after peripheral nerve injury activates IRF5. IRF5 specifically and selectively binds to the promoter of the purinergic receptor P2X 4 (P2RX4) gene encoding the receptor P2X4R, which is expressed on the cell membrane of microglia [36,38-41]. Activated P2X4 receptors bind adenosine triphosphate (ATP) and lead to signaling (activation) of other pro-inflammatory pathways and maintenance of neuropathic pain [40,42,43]. CSF-1 is now considered one of the factors that end up in the spinal cord and are associated with the activation of microglia. Both CSF-1 and DAP12 are important pharmacologic targets.
The fractalkine receptor, C-X3-C motif chemokine receptor 1 (CX3CR1), participates in microglial activation as well. This receptor is expressed on the cell membrane of microglial cells of the spinal cord and brain, it binds to fractalkine (chemokine-adhesion molecule) and is overexpressed in the spinal cord during inflammation and nerve damage [44,45]. The binding of fractalkine to its receptor (CX3CR1) initiates the p38/MAPK serine/threonine protein kinase intracellular signaling pathway. This is considered the most basic signaling pathway inside microglia that controls cell proliferation, cell response to stress, cell death, differentiation, migration, etc. It is associated with the synthesis and release of a wide variety of cellular mediators of inflammation, such as brain-derived neurotrophic factor (BDNF), TNF-α, IL-1b, and IL-6 by microglia, which, when secreted, enhance synaptic transmission and stimulate interstitial facilitatory neurons [41,46-48].
Summarizing, three main elements could be reported in the mechanism of activation of the spinal microglia. First, nerve damage leads to de novo CSF-1 induction in the dorsal root ganglion. From here, CSF-1 reaches the ends of the sensory neuron (reciprocating movement) and recruits the CSF-1 receptors (CSF-1Rs). Second, the presence of CSF-1 is considered a capable and necessary condition for the proliferation, migration, and activation of microglia. Finally, the activation simultaneously affects the entire reflex arc, the sensory neurons of the dorsal horn, the medial neurons, and the corresponding anterior horn motor neurons. The purine (ATP) and fractalkine pathways are rather involved in the maintenance and perpetuation of pain than in the initial activation and sensitization.
Activation of the spinal neuroglia is observed in the initial phase (onset phase) of neuropathic pain and precedes the activation of astrocytes (astrogliosis), supporting the hypothesis that microglia contribute to the initiation of neuropathic pain while astrocytes participate in its maintenance [49,50]. There is considerable evidence that microglial activation is both a sufficient and necessary condition to produce central sensitization and neuropathic pain [51,52].
Events in the brain
The role of neurotransmitters in the function of microglia has been examined in the brain. Neuropeptide Y (NPY), vasoactive intestinal polypeptide (VIP), pituitary adenylate cyclase-activating peptide (PACAP), somatostatin, cortistatin, tachykinins, calcitonin gene-related peptide (CGRP), leptin, ghrelin, and neuropeptides derived from the POMC gene (proopiomelanocortin - hypothalamic nucleus toxoid), such as adenocorticotropin-releasing hormone (ACTH), melanocyte-stimulating hormone (MSH), and opioid peptides (μ, δ, and κ receptors) directly affect the function of microglial cells. Microglial cells express membrane receptors for all the above-mentioned neuropeptides, thus participating in the regulation of pain sensitivity [53,54].
The role of brain microglia in chronic neuropathic pain also emerged from animal studies [55,56]. It was found that peripheral nerve damage led to significant microglial activation (microgliosis) in the thalamus, somatosensory cortex, and parietal system, brain areas associated with chronic pain, and the emotional and cognitive response to it [57]. The same research group had reported a year before [58] that a peripheral nerve lesion activates microglia selectively and focally in the region of the mesolimbic dopaminergic system, affecting the entire reward circuit [59-61].
Targeting microglia as a therapeutic strategy in treating neuropathic pain
Since the involvement of microglia in the development and maintenance of neuropathic pain has been confirmed, the investigation of new drugs able to modify the function of immune cells of the CNS is a priority and a challenge for researchers [62]. There is an urgent need for new, safe, and effective drugs for neuropathic pain, which could constitute a combination therapy that targets both neural (neurons) and non-neural (microglia and astrocytes) cells. Since microglial cells are immune cells of the CNS, they would be suppressed using anti-inflammatory and immunosuppressive drugs. Both drug categories have been used to reduce microglia activation, control inflammation and neuroinflammation, and improve the clinical symptoms of neuropathic pain [63]. Table 1 summarizes the immunomodulatory drugs that target microglia and potentially may be used to treat neuropathic pain, according to animal and experimental studies.
Table 1. Anti-inflammatory and immunomodulatory drugs that target microglial cells.
MAP: mitogen-activated protein TNF: tumor necrosis factor CNS: central nervous system NMDA: N-methyl-D-aspartate IL: interleukin COX: cyclooxygenase PPAR: peroxisome proliferator-activated receptor
| Drug/substance | Mechanism of action | Effect on models of neuropathic pain | References |
| Minocycline | Tetracycline. Partly immunosuppressive because it inhibits the p38 MAP signaling pathway and cellular growth | Prevents the development of neuropathic pain (hyperalgesia and allodynia) by inhibiting microglial cells when given before the nerve injury | [1,51,59,64-70] |
| Pentoxifylline | Non-selective phosphodiesterase inhibitor. Potent TNF-α inhibitor | Reduces mechanical allodynia by suppressing the activation of microglia and astrocytes. It is often accompanied by a decrease in pro-inflammatory cytokines | [22,27,65,69,71,72] |
| Methotrexate | Reduces the concentration of folic acid, de novo inhibits purines and synthesis of thymidylic acid | Inhibits the activation and proliferation of microglia. Effective when administered after nerve damage | [73] |
| Nucleotide receptor antagonists | The activation of P2X & P2Y purinergic receptors modulates the activity of peripheral immune cells and microglial cells in the CNS | Inhibit the activation of peripheral macrophages and spinal microglia | [74] |
| P38 MAP kinase inhibitors and ketamine | Inhibit important signaling pathways of microglia. Specifically, ketamine inhibits NMDA receptors | Reduce tactile allodynia. Effective when administered before nerve damage. Use of ketamine in neuropathic pain. | [50,51,75-77] |
| Neutralizing antibodies and receptor – trapping strategies | Substances that control the synthesis of cytokines and act against IL1, IL6, IL10, & TNF | They reduce the biological effects of pro-inflammatory cytokines. IL10 also has anti-inflammatory action | [69] |
| Complement inhibitors | They prevent the activation of the complement expressed by microglia. They block the classical pathway of the complement system | Inhibit complement activation pathways, including the C5 pathway, which acts as a chemoattractant in the posterior horn of the spinal cord | [69,78] |
| Cannabinoids | Activate CB1 & CB2 cannabinoid receptors, which are particularly expressed in microglia | CB2 receptors regulate the migration and proliferation of immune cells. CB2 agonists reduce mechanical allodynia | [79,80] |
| Nonsteroidal anti-inflammatory drugs | Inhibition of prostaglandin PGE2 of microglial cells, inhibition of COX, inactivation of microglial via PPAR-γ pathway | Anti-inflammatory action. Inhibition of activated microglia. Inhibition of β-amyloid production in the brain, inhibition of β and γ-secretase. Reduction of proliferation | [13,79,81-84] |
Conclusions
Peripheral nerve damage and peripheral inflammation are known causes of chronic pain. In situations of sensitization, prolonged activation of the primary sensory nerves in the periphery is followed by increased activity of the terminal nerve cells in the posterior horn of the spinal cord. However, hyperactivity of the nerves in the area has the effect of activating non-nerve cells (immune cells) at the site of the lesion (partial activation). Microglial cells are thus involved in nervous pathology (disease or injury) dynamically; they proliferate, change morphologically, and secrete substances that make local synapses even more sensitive. Therefore, a condition of positive stimulus feedback is established, leading to chronicity. Activation of microglia is more likely after nerve damage than after inflammation. They are found along the entire length of the somatosensory pathway, where immune cells are present at each synapsis. Microglia participate in all processes of the mechanism that produces chronic neuropathic pain (gliopathy). The interaction between neurons and microglial cells is considered critical for chronic pain and inflammation. The disruption of the two-way relationship between neurons and immune cells is associated not only with chronic neuropathic pain but is considered the pathogenetic factor for a number of inflammatory diseases of the peripheral and central nervous system, which are called neurodegenerative diseases.
Concluding, the somatosensory system is a key regulator of the innate immune response of organisms. To achieve this, it interacts and cooperates with the cells of the immune system, in particular with microglia. Understanding this communication, which we have tried to describe in detail in this review, will unfold new cellular mechanisms, lead research into interesting biochemical molecular pathways, and turn medical thought to a more holistic view of the organic systems involved in chronic pain (neuro-immune-endocrine coupling).
The authors have declared that no competing interests exist.
References
- 1.Neuropathic pain: a maladaptive response of the nervous system to damage. Costigan M, Scholz J, Woolf CJ. Annu Rev Neurosci. 2009;32:1–32. doi: 10.1146/annurev.neuro.051508.135531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Diagnosis and management of neuropathic pain: review of literature and recommendations of the Polish Association for the Study of Pain and the Polish Neurological Society - Part two. Szczudlik A, Dobrogowski J, Wordliczek J, et al. Neurol Neurochir Pol. 2014;48:423–435. doi: 10.1016/j.pjnns.2014.11.002. [DOI] [PubMed] [Google Scholar]
- 3.The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. Austin PJ, Moalem-Taylor G. J Neuroimmunol. 2010;229:26–50. doi: 10.1016/j.jneuroim.2010.08.013. [DOI] [PubMed] [Google Scholar]
- 4.Therapeutic management of chronic neuropathic pain: an examination of pharmacologic treatment. Vadalouca A, Siafaka I, Argyra E, Vrachnou E, Moka E. Ann N Y Acad Sci. 2006;1088:164–186. doi: 10.1196/annals.1366.016. [DOI] [PubMed] [Google Scholar]
- 5.Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Finnerup NB, Attal N, Haroutounian S, et al. Lancet Neurol. 2015;14:162–173. doi: 10.1016/S1474-4422(14)70251-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Microglia: new roles for the synaptic stripper. Kettenmann H, Kirchhoff F, Verkhratsky A. Neuron. 2013;77:10–18. doi: 10.1016/j.neuron.2012.12.023. [DOI] [PubMed] [Google Scholar]
- 7.Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Elmore MR, Najafi AR, Koike MA, et al. Neuron. 2014;82:380–397. doi: 10.1016/j.neuron.2014.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. J Neurosci. 2009;29:3974–3980. doi: 10.1523/JNEUROSCI.4363-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Contributions of microglia to structural synaptic plasticity. Kim KH, Son SM, Mook-Jung I. J Exp Neurosci. 2013;7:85–91. doi: 10.4137/JEN.S11269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.What do microglia really do in healthy adult brain? Augusto-Oliveira M, Arrifano GP, Lopes-Araújo A, et al. Cells. 2019;8 doi: 10.3390/cells8101293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Neuropathic pain: biomolecular intervention and imaging via targeting microglia activation. Ji A, Xu J. Biomolecules. 2021;11 doi: 10.3390/biom11091343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.The role of glia and the immune system in the development and maintenance of neuropathic pain. Vallejo R, Tilley DM, Vogel L, Benyamin R. Pain Pract. 2010;10:167–184. doi: 10.1111/j.1533-2500.2010.00367.x. [DOI] [PubMed] [Google Scholar]
- 13.The role of microglia in the pathobiology of neuropathic pain development: what do we know? Zhao H, Alam A, Chen Q, et al. Br J Anaesth. 2017;118:504–516. doi: 10.1093/bja/aex006. [DOI] [PubMed] [Google Scholar]
- 14.Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Inoue K, Tsuda M. Nat Rev Neurosci. 2018;19:138–152. doi: 10.1038/nrn.2018.2. [DOI] [PubMed] [Google Scholar]
- 15.Intraspinal non-neuronal cellular responses to peripheral nerve injury. Gilmore SA, Skinner RD. Anat Rec. 1979;194:369–387. doi: 10.1002/ar.1091940305. [DOI] [PubMed] [Google Scholar]
- 16.Perspectives in pain research 2014: neuroinflammation and glial cell activation: the cause of transition from acute to chronic pain? Cairns BE, Arendt-Nielsen L, Sacerdote P. Scand J Pain. 2015;6:3–6. doi: 10.1016/j.sjpain.2014.10.002. [DOI] [PubMed] [Google Scholar]
- 17.Neuroinflammation and glial cell activation in pathogenesis of chronic pain. Hansson E. Scand J Pain. 2015;6:1–2. doi: 10.1016/j.sjpain.2014.10.004. [DOI] [PubMed] [Google Scholar]
- 18.Microglia: sculptors of neuropathic pain? Ward H, West SJ. R Soc Open Sci. 2020;7:200260. doi: 10.1098/rsos.200260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Do glial cells control pain? Suter MR, Wen YR, Decosterd I, Ji RR. Neuron Glia Biol. 2007;3:255–268. doi: 10.1017/S1740925X08000100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Yi MH, Liu YU, Liu K, et al. Brain Behav Immun. 2021;92:78–89. doi: 10.1016/j.bbi.2020.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. Nature. 2003;424:778–783. doi: 10.1038/nature01786. [DOI] [PubMed] [Google Scholar]
- 22.Neuropathic pain: mechanisms and their clinical implications. Cohen SP, Mao J. BMJ. 2014;348:0. doi: 10.1136/bmj.f7656. [DOI] [PubMed] [Google Scholar]
- 23.Nociceptor sensory neuron-immune interactions in pain and inflammation. Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Trends Immunol. 2017;38:5–19. doi: 10.1016/j.it.2016.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Neurotransmitter receptors on microglia. Liu H, Leak RK, Hu X. Stroke Vasc Neurol. 2016;1:52–58. doi: 10.1136/svn-2016-000012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Resting microglial motility is independent of synaptic plasticity in mammalian brain. Wu LJ, Zhuo M. J Neurophysiol. 2008;99:2026–2032. doi: 10.1152/jn.01210.2007. [DOI] [PubMed] [Google Scholar]
- 26.Spinal microglial motility is independent of neuronal activity and plasticity in adult mice. Chen T, Koga K, Li XY, Zhuo M. Mol Pain. 2010;6:19. doi: 10.1186/1744-8069-6-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Neuroinflammation and the generation of neuropathic pain. Ellis A, Bennett DL. Br J Anaesth. 2013;111:26–37. doi: 10.1093/bja/aet128. [DOI] [PubMed] [Google Scholar]
- 28.The “missing link” in the physiology of pain: glial cells. Peppin JF, Raffa RB. https://www.practicalpainmanagement.com/treatments/missing-link-physiology-pain-glial-cells Pract Pain Manag. 2016;16 [Google Scholar]
- 29.Microglia in pain: detrimental and protective roles in pathogenesis and resolution of pain. Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR. Neuron. 2018;100:1292–1311. doi: 10.1016/j.neuron.2018.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Microglia in the spinal cord and neuropathic pain. Tsuda M. J Diabetes Investig. 2016;7:17–26. doi: 10.1111/jdi.12379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Microglia in the CNS and Neuropathic Pain. Tsuda M. Adv Exp Med Biol. 2018;1099:77–91. doi: 10.1007/978-981-13-1756-9_7. [DOI] [PubMed] [Google Scholar]
- 32.Neuropathic pain and spinal microglia: a big problem from molecules in "small" glia. Tsuda M, Inoue K, Salter MW. Trends Neurosci. 2005;28:101–107. doi: 10.1016/j.tins.2004.12.002. [DOI] [PubMed] [Google Scholar]
- 33.Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain. Coull JA, Boudreau D, Bachand K, et al. Nature. 2003;424:938–942. doi: 10.1038/nature01868. [DOI] [PubMed] [Google Scholar]
- 34.Peripheral nerve injury and TRPV1-expressing primary afferent C-fibers cause opening of the blood-brain barrier. Beggs S, Liu XJ, Kwan C, Salter MW. Mol Pain. 2010;6:74. doi: 10.1186/1744-8069-6-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.P2X4R+ microglia drive neuropathic pain. Beggs S, Trang T, Salter MW. Nat Neurosci. 2012;15:1068–1073. doi: 10.1038/nn.3155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.The known knowns of microglia-neuronal signalling in neuropathic pain. Beggs S, Salter MW. Neurosci Lett. 2013;557 Pt A:37–42. doi: 10.1016/j.neulet.2013.08.037. [DOI] [PubMed] [Google Scholar]
- 37.Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Guan Z, Kuhn JA, Wang X, et al. Nat Neurosci. 2016;19:94–101. doi: 10.1038/nn.4189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Snapshot: microglia in disease. Beggs S, Salter MW. Cell. 2016;165:1294. doi: 10.1016/j.cell.2016.05.036. [DOI] [PubMed] [Google Scholar]
- 39.Molecules in pain and sex: a developing story. Mapplebeck JC, Beggs S, Salter MW. Mol Brain. 2017;10:9. doi: 10.1186/s13041-017-0289-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Transcription factor IRF5 drives P2X4R+-reactive microglia gating neuropathic pain. Masuda T, Iwamoto S, Yoshinaga R, et al. Nat Commun. 2014;5:3771. doi: 10.1038/ncomms4771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.IRF8 is a critical transcription factor for transforming microglia into a reactive phenotype. Masuda T, Tsuda M, Yoshinaga R, Tozaki-Saitoh H, Ozato K, Tamura T, Inoue K. Cell Rep. 2012;1:334–340. doi: 10.1016/j.celrep.2012.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Microglia-mediated regulation of neuropathic pain: molecular and cellular mechanisms. Tsuda M. Biol Pharm Bull. 2019;42:1959–1968. doi: 10.1248/bpb.b19-00715. [DOI] [PubMed] [Google Scholar]
- 43.P2X4 receptors and neuropathic pain. Tsuda M, Masuda T, Tozaki-Saitoh H, Inoue K. Front Cell Neurosci. 2013;7:191. doi: 10.3389/fncel.2013.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Multiple P2Y subtypes in spinal microglia are involved in neuropathic pain after peripheral nerve injury. Kobayashi K, Yamanaka H, Yanamoto F, Okubo M, Noguchi K. Glia. 2012;60:1529–1539. doi: 10.1002/glia.22373. [DOI] [PubMed] [Google Scholar]
- 45.Genetically determined P2X7 receptor pore formation regulates variability in chronic pain sensitivity. Sorge RE, Trang T, Dorfman R, et al. Nat Med. 2012;18:595–599. doi: 10.1038/nm.2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Glia and pain: is chronic pain a gliopathy? Ji RR, Berta T, Nedergaard M. Pain. 2013;154 Suppl 1:0–28. doi: 10.1016/j.pain.2013.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Kawasaki Y, Xu ZZ, Wang X, et al. Nat Med. 2008;14:331–336. doi: 10.1038/nm1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Microglia-neuron interactions in the models of neuropathic pain. Tozaki-Saitoh H, Tsuda M. Biochem Pharmacol. 2019;169:113614. doi: 10.1016/j.bcp.2019.08.016. [DOI] [PubMed] [Google Scholar]
- 49.Glial cells and chronic pain. Gosselin RD, Suter MR, Ji RR, Decosterd I. Neuroscientist. 2010;16:519–531. doi: 10.1177/1073858409360822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.p38 MAPK, microglial signaling, and neuropathic pain. Ji RR, Suter MR. Mol Pain. 2007;3:33. doi: 10.1186/1744-8069-3-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Targeting the microglial signaling pathways: new insights in the modulation of neuropathic pain. Popiolek-Barczyk K, Mika J. Curr Med Chem. 2016;23:2908–2928. doi: 10.2174/0929867323666160607120124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Microglia and spinal cord synaptic plasticity in persistent pain. Taves S, Berta T, Chen G, Ji RR. Neural Plast. 2013;2013:753656. doi: 10.1155/2013/753656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Neuropeptides and microglial activation in inflammation, pain, and neurodegenerative diseases. Carniglia L, Ramírez D, Durand D, et al. Mediators Inflamm. 2017;2017:5048616. doi: 10.1155/2017/5048616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Change pain: Ever evolving-an update for 2016. Pergolizzi JV, Paladini A, Varrassi G, Raffa RB. Pain Ther. 2016;5:127–133. doi: 10.1007/s40122-016-0058-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Topography of microglial activation in sensory- and affect-related brain regions in chronic pain. Taylor AM, Mehrabani S, Liu S, Taylor AJ, Cahill CM. J Neurosci Res. 2017;95:1330–1335. doi: 10.1002/jnr.23883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Correlation between ventral striatal catecholamine content and nociceptive thresholds in neuropathic mice. Taylor AM, Murphy NP, Evans CJ, Cahill CM. J Pain. 2014;15:878–885. doi: 10.1016/j.jpain.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: sex-dependent microglial signaling in the spinal cord. Taves S, Berta T, Liu DL, et al. Brain Behav Immun. 2016;55:70–81. doi: 10.1016/j.bbi.2015.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Microglia disrupt mesolimbic reward circuitry in chronic pain. Taylor AM, Castonguay A, Taylor AJ, et al. J Neurosci. 2015;35:8442–8450. doi: 10.1523/JNEUROSCI.4036-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Evidence for brain glial activation in chronic pain patients. Loggia ML, Chonde DB, Akeju O, et al. Brain. 2015;138:604–615. doi: 10.1093/brain/awu377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Neurons and Microglia; A Sickly-Sweet Duo in Diabetic Pain Neuropathy. Rajchgot T, Thomas SC, Wang JC, et al. Front Neurosci. 2019;13:25. doi: 10.3389/fnins.2019.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Key role for spinal dorsal horn microglial kinin B1 receptor in early diabetic pain neuropathy. Talbot S, Chahmi E, Dias JP, Couture R. J Neuroinflammation. 2010;7:36. doi: 10.1186/1742-2094-7-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Microglia and chronic pain. Malcangio M. Pain. 2016;157:1002–1003. doi: 10.1097/j.pain.0000000000000353. [DOI] [PubMed] [Google Scholar]
- 63.Microglial TNFα induces COX2 and PGI2 synthase expression in spinal endothelial cells during neuropathic pain. Kanda H, Kobayashi K, Yamanaka H, Okubo M, Noguchi K. eNeuro. 2017;4 doi: 10.1523/ENEURO.0064-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Activated microglia contribute to the maintenance of chronic pain after spinal cord injury. Hains BC, Waxman SG. J Neurosci. 2006;26:4308–4317. doi: 10.1523/JNEUROSCI.0003-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.The neuropathic pain triad: neurons, immune cells and glia. Scholz J, Woolf CJ. Nat Neurosci. 2007;10:1361–1368. doi: 10.1038/nn1992. [DOI] [PubMed] [Google Scholar]
- 66.Neuron-glia crosstalk and neuropathic pain: involvement in the modulation of motor activity in the orofacial region. Hossain MZ, Unno S, Ando H, Masuda Y, Kitagawa J. Int J Mol Sci. 2017;18 doi: 10.3390/ijms18102051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Reduced spinal microglial activation and neuropathic pain after nerve injury in mice lacking all three nitric oxide synthases. Kuboyama K, Tsuda M, Tsutsui M, et al. Mol Pain. 2011;7:50. doi: 10.1186/1744-8069-7-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Microglia P2X4 receptor contributes to central sensitization following recurrent nitroglycerin stimulation. Long T, He W, Pan Q, et al. J Neuroinflammation. 2018;15:245. doi: 10.1186/s12974-018-1285-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Mika J. https://pubmed.ncbi.nlm.nih.gov/18622054/ Pharmacol Rep. 2008;60:297–307. [PubMed] [Google Scholar]
- 70.Age-related differences in neuropathic pain behavior and spinal microglial activity after L5 spinal nerve ligation in male rats. Zeinali H, Manaheji H, Zaringhalam J, Bahari Z, Nazemi S, Sadeghi M. Basic Clin Neurosci. 2016;7:203–212. doi: 10.15412/J.BCN.03070305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Microglial signaling in chronic pain with a special focus on caspase 6, p38 MAP kinase, and sex dependence. Berta T, Qadri YJ, Chen G, Ji RR. J Dent Res. 2016;95:1124–1131. doi: 10.1177/0022034516653604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pathological and protective roles of glia in chronic pain. Milligan ED, Watkins LR. Nat Rev Neurosci. 2009;10:23–36. doi: 10.1038/nrn2533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Low-dose methotrexate reduces peripheral nerve injury-evoked spinal microglial activation and neuropathic pain behavior in rats. Scholz J, Abele A, Marian C, Häussler A, Herbert TA, Woolf CJ, Tegeder I. Pain. 2008;138:130–142. doi: 10.1016/j.pain.2007.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.The role of nucleotides in the neuron—glia communication responsible for the brain functions. Inoue K, Koizumi S, Tsuda M. J Neurochem. 2007;102:1447–1458. doi: 10.1111/j.1471-4159.2007.04824.x. [DOI] [PubMed] [Google Scholar]
- 75.Nerve injury and neuropathic pain - a question of age. Fitzgerald M, McKelvey R. Exp Neurol. 2016;275 Pt 2:296–302. doi: 10.1016/j.expneurol.2015.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Non-specific low back pain. Maher C, Underwood M, Buchbinder R. Lancet. 2017;389:736–747. doi: 10.1016/S0140-6736(16)30970-9. [DOI] [PubMed] [Google Scholar]
- 77.Ketamine for chronic pain: risks and benefits. Niesters M, Martini C, Dahan A. Br J Clin Pharmacol. 2014;77:357–367. doi: 10.1111/bcp.12094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Complement induction in spinal cord microglia results in anaphylatoxin C5a-mediated pain hypersensitivity. Griffin RS, Costigan M, Brenner GJ, et al. J Neurosci. 2007;27:8699–8708. doi: 10.1523/JNEUROSCI.2018-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Moulin D, Boulanger A, Clark AJ, et al. Pain Res Manag. 2014;19:328–335. doi: 10.1155/2014/754693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Neuroimmune interactions and pain: focus on glial-modulating targets. Romero-Sandoval EA, Horvath RJ, DeLeo JA. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2696046/ Curr Opin Investig Drugs. 2008;9:726–734. [PMC free article] [PubMed] [Google Scholar]
- 81.Non-steroidal anti-inflammatory drugs and brain inflammation: effects on microglial functions. Ajmone-Cat MA, Bernardo A, Greco A, Minghetti L. Pharmaceuticals (Basel) 2010;3:1949–1965. doi: 10.3390/ph3061949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer's disease in rodent models. Daniels MJ, Rivers-Auty J, Schilling T, et al. Nat Commun. 2016;7:12504. doi: 10.1038/ncomms12504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Pain regulation by non-neuronal cells and inflammation. Ji RR, Chamessian A, Zhang YQ. Science. 2016;354:572–577. doi: 10.1126/science.aaf8924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Prostaglandin E2 E‐prostanoid 4 receptor signaling in microglia reduces inflammation and amyloid burden in a mouse model of Alzheimer's disease. Alzheimer's & dementia. Woodling N, Shi J, Priyam P, Wang Q, Andreasson K. 2011;7:12. [Google Scholar]
