Chronic pain is maintained in part by central sensitization, a phenomenon of synaptic plasticity, and increased neuronal responsiveness in central pain pathways after painful insults. Accumulating evidence suggests that central sensitization is also driven by neuroinflammation in the peripheral and central nervous system. A characteristic feature of neuroinflammation is the activation of glial cells, such as microglia and astrocytes, in the spinal cord and brain, leading to the release of proinflammatory cytokines and chemokines. Recent studies suggest that central cytokines and chemokines are powerful neuromodulators and play a sufficient role in inducing hyperalgesia and allodynia after central nervous system administration. Sustained increase of cytokines and chemokines in the central nervous system also promotes chronic widespread pain that affects multiple body sites. Thus, neuroinflammation drives widespread chronic pain via central sensitization. We also discuss sex-dependent glial/immune signaling in chronic pain and new therapeutic approaches that control neuroinflammation for the resolution of chronic pain.

Chronic pain is a major health concern that affects one in three Americans and costs the U.S. economy $635 billion dollars each year.1,2  Acute pain is often elicited by acute inflammation and has biologic significance to protect the wounded tissue. Chronic pain is maladaptive, has no beneficial biologic significance, and is characterized by spontaneous pain (e.g., burning) as well as evoked pain in response to noxious (hyperalgesia) or nonnoxious (allodynia) stimuli. It is generally believed that neuronal plasticity in pain-coding pathways and circuits results in chronic pain. Neuronal plasticity consists of peripheral sensitization in primary sensory neurons of dorsal root ganglia and trigeminal ganglia3–5  and central sensitization of pain-processing neurons in the spinal cord and brain.6–9 

The perception of pain is typically associated with inflammation, a complex biologic response of the somatosensory, immune, neuronal, autonomic, and vascular/circulatory system to tissue damage, pathogens, or irritants. Acute inflammation, which generally results in perception of pain, serves an important protective or survival role by removing harmful stimuli, initiating the healing process, and restoring tissue integrity (table 1). Primary afferents that respond to tissue injury (i.e., nociceptors) include unmyelinated C-fibers and myelinated Aδ-fibers that terminate in skin, muscle, joints, and visceral organs, with their cell bodies located in dorsal root ganglia and trigeminal ganglia. Nociceptors are activated or sensitized by inflammatory mediators such as bradykinin, prostaglandins, nerve growth factor, and proinflammatory cytokines such as tumor necrosis factor-α, interleukin 1β, and proinflammatory chemokines (e.g., [CC motif] ligand 2, CXC motif chemokine 5)3,10,11  that directly bind and stimulate G-protein–coupled receptors, ionotropic receptors, and tyrosine kinase receptors. It is noteworthy that all of these receptors are expressed on the terminals and/or cell bodies of nociceptors.3,5  Cytokine profiles observed in human skin are also associated with inflammation and pain and thus may serve as biomarkers for chronic pain.12,13  Activation of a mosaic of peripheral receptors results in hypersensitivity and hyperexcitability of nociceptor neurons (peripheral sensitization) through modulation of various ion channels, such as transient receptor potential ion channels (e.g., transient receptor potential ion channels A1, V1, and V4), sodium channels (e.g., subtypes Nav1.7/1.8/1.9),3,4,14  and mechanosensitive Piezo ion channels.15,16  MicroRNA may serve as a novel inflammatory and pain-evoking mediator. For example, miR-let-7b induces spontaneous pain via activation of Toll-like receptor 7 and transient receptor potential ion channel A1.17  Furthermore, the activation of protein kinases such as mitogen-activated protein kinases, protein kinase A, and protein kinase C in primary sensory neurons critically contributes to the induction and maintenance of peripheral sensitization. For instance, activation of p38 mitogen-activated protein kinase in dorsal root ganglia neurons initiates peripheral sensitization by increasing transient receptor potential ion channel V1 activity in response to tumor necrosis factor18  and also by increasing NaV1.8 activity in response to interleukin-1β, and furthermore, this activation maintains peripheral sensitization and chronic pain by increasing transient receptor potential ion channel V1 expression.20,21  In parallel, peripheral tumor necrosis factor and interleukin-1β have been strongly implicated in the pathogenesis of inflammatory and neuropathic pain.22–26 

Table 1.

Comparison of Inflammation, Neurogenic Inflammation, and Neuroinflammation with Special Focus on Location, Features, and Role in Pain

Comparison of Inflammation, Neurogenic Inflammation, and Neuroinflammation with Special Focus on Location, Features, and Role in Pain
Comparison of Inflammation, Neurogenic Inflammation, and Neuroinflammation with Special Focus on Location, Features, and Role in Pain

Of note, nociceptors and immune cells have bidirectional interactions.27  Nociceptors not only listen to immune cells by responding to inflammatory mediators but also talk to immune cells and modulate the immune response to inflammation.28,29  Like immune cells, nociceptors express cytokines, chemokines, and Toll-like receptors that are essential for immune modulation.19,30–32  Release of cytokines and chemokines from nociceptors can rapidly regulate resident immune cells and attract circulating cells to the area of local inflammation that engage primary afferents and cell bodies in the nerve and dorsal root ganglia. For example, nociceptor-produced chemokine (CC motif) ligand 2 regulates local macrophage activation in dorsal root ganglia after chemotherapy via Toll-like receptor signaling, resulting in neuropathic pain.19,33  Activation of nociceptors, especially C-fibers, also produces neurogenic inflammation via releasing neuropeptides such as substance P and calcitonin gene–related peptide or prostanoids28  (table 1). Neurogenic inflammation occurs immediately after intradermal administration of capsaicin and mustard oil via respective activation of transient receptor potential ion channels A1 and V1. Neurogenic inflammation results in rapid plasma extravasation and edema, even before the infiltration of immune cells. Neurogenic inflammation plays an important role in inflammatory diseases such as asthma and psoriasis but also contributes to pain conditions such as migraine and complex regional pain syndrome after bone fracture.34  Nociceptors may differentially regulate inflammation in a context-dependent manner. For example, ablation of nociceptors decreases neurogenic inflammation but also enhances inflammation after bacterial infection by releasing calcitonin gene–related peptide.35 

Peripheral inflammation with resulting persistent nociceptive input also leads to the increased release of neurotransmitters (glutamate, substance P, calcitonin gene–related peptide, and brain-derived growth factor) from the primary afferent central terminals in the spinal cord and trigeminal nucleus. Through signal transduction, these neurotransmitters produce a state of neuronal hyperactivity and hyperexcitability in the spinal cord and brain known as central sensitization.36,37  Activation of postsynaptic glutamate N-methyl-d-aspartate (NMDA) receptors and plasma cell membrane surface insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are essential steps for the induction and maintenance of central sensitization.6,38 

A PubMed Search on September 30, 2017, using the keywords “neuroinflammation and pain” reveals a substantial increase in the number of publications in the last 10 yr, jumping from 12 in 2008 to 150 in 2016. There is also a similar increase in the number of “microglia and pain” publications over the same period of time (fig. 1). Neuroinflammation is a localized form of inflammation that occurs in the peripheral nervous system (including peripheral nerves and ganglia) and central nervous system (CNS, including the spinal cord and brain). Characteristic features of neuroinflammation are (1) vasculature changes that result in increased vascular permeability, (2) infiltration of leukocytes, (3) activation of glial cells, and (4) production of inflammatory mediators including cytokines and chemokines (table 1). Although the CNS is normally protected by the blood–brain barrier, increased permeability of the blood–brain barrier is an important feature of neuroinflammation, leading to increased leukocyte invasion to the CNS. It is becoming increasingly appreciated that neuroinflammation is a major cause of several neurologic and neuropsychiatric diseases such as Alzheimer disease, Parkinson disease, multiple sclerosis, depression, bipolar disorder, and autism, as well as postoperative complications like neurocognitive disorders (e.g., delirium).39–41 

Fig. 1.

A PubMed Search on September 30, 2017, shows the number of publications on “neuroinflammation and pain” and “microglia and pain” in the last 10 yr. The numbers at the bottom show the total number of publications in PubMed.

Fig. 1.

A PubMed Search on September 30, 2017, shows the number of publications on “neuroinflammation and pain” and “microglia and pain” in the last 10 yr. The numbers at the bottom show the total number of publications in PubMed.

Close modal

It is noteworthy that neuroinflammation is associated with various painful insults and pathologies (fig. 2),39  including but not limited to trauma such as traumatic brain injury, stroke, spinal cord injury, major surgeries including both cardiac and noncardiac procedures (e.g., amputation, thoracotomy, and mastectomy), and autoimmune diseases (rheumatoid arthritis and multiple sclerosis). Other painful conditions, such as osteoarthritis, peripheral cancers (e.g., bone cancers), and viral infections (e.g., shingles/herpes zoster by varicella-zoster virus) also induce neuroinflammation in the peripheral nervous system and CNS.27,42  Furthermore, painful neuroinflammation (e.g., glial activation) can be induced by chemotherapy drugs such as paclitaxel, anti–human immunodeficiency virus (HIV) treatment, and chronic opioid treatment.43–46  Immune therapies are increasingly appreciated for treating cancers by boosting the activities of immune cells such as T cells and other immune cells.47,48  These therapies also change pain sensitivity by modulating inflammation and neuroinflammation. Deficiency of the negative immune regulator B7-H1, also called programmed death ligand 1, enhances inflammation and neuropathic pain after chronic constriction injury of mouse sciatic nerve.49  Notably, PD-1, the receptor for programmed death ligand 1, is not only present in immune cells but also expressed in nociceptor neurons of mouse and human dorsal root ganglia. Furthermore, programmed death ligand 1 can potently suppress nociceptor activities via regulating sodium and potassium ion channels in mouse and human nociceptors.50  Many of these painful insults involve brain and spinal trauma and nerve injury resulting from major surgeries, drug treatments, and diabetic neuropathy in neuropathic pain conditions. Neurogenic inflammation of the skin and joints is not only induced by activation of peripheral C-fibers but may also be triggered by the dorsal root reflex in the spinal cord after either orthograde or anterograde neuronal activation, which permits peripheral inflammatory responses to occur via local insults or by CNS “top-down” activation of primary afferents.51,52  Furthermore, neuroinflammation in the spinal cord and brain can also be neurogenic after neuronal activation in the CNS.53 

Fig. 2.

Neuroinflammation is associated with various insults that evoke painful sensations. These insults include but are not limited to trauma, major surgeries, drug treatments, autoimmune disease conditions, and other painful insults and tissue damage. Some of these insults such as major surgeries (breast surgery, amputation, thoracotomy), chemotherapy, and antiviral treatment will cause nerve injury, as highlighted in red. Others will cause immune activation (highlighted in purple) and tissue injury (highlighted in light blue). Neuroinflammation results in several adverse effects, such as chronic pain and neurodegenerative diseases including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), and stroke. Neuroinflammation is also associated with chronic overlapping pain conditions. After priming of the nociceptive circuit by previous injury, stress, or existing genomic, environmental, and psychologic factors, acute insult may cause transition from acute pain to chronic pain. CNS = central nervous system.

Fig. 2.

Neuroinflammation is associated with various insults that evoke painful sensations. These insults include but are not limited to trauma, major surgeries, drug treatments, autoimmune disease conditions, and other painful insults and tissue damage. Some of these insults such as major surgeries (breast surgery, amputation, thoracotomy), chemotherapy, and antiviral treatment will cause nerve injury, as highlighted in red. Others will cause immune activation (highlighted in purple) and tissue injury (highlighted in light blue). Neuroinflammation results in several adverse effects, such as chronic pain and neurodegenerative diseases including Alzheimer disease (AD), Parkinson disease (PD), multiple sclerosis (MS), and stroke. Neuroinflammation is also associated with chronic overlapping pain conditions. After priming of the nociceptive circuit by previous injury, stress, or existing genomic, environmental, and psychologic factors, acute insult may cause transition from acute pain to chronic pain. CNS = central nervous system.

Close modal

Although it is widely believed that chronic pain persists after the observable signs and symptoms of inflammation have resolved,54  our understanding of neuroinflammation is changing this perspective. We now recognize that neuroinflammation is associated with and perhaps mediates the persistence and chronification of human pain conditions. Given the close proximity to pain neurocircuits, mediators of neuroinflammation are highly effective in modulating pain sensitivity. In particular, inflammation and neuroinflammation are differentially correlated with chronic pain. For example, HIV-infected patients with neuropathic pain show permanent neuroinflammation in the spinal cord.55  Patients with fibromyalgia, a chronic widespread musculoskeletal pain condition,56  also exhibit small fiber neuropathy together with chronic neuroinflammation.57  The degree to which acute and chronic pain perception is mediated by the same cast of neuroinflammatory mediators is an interesting and open question.

It is noteworthy that a recent study by Wei et al.58  has shown distinct roles for inflammation and central neuroinflammation in the acute phase versus the chronic phase of pain behavior in a rat model of complex regional pain syndrome. Although peripheral interleukin-1β levels only increased at 4 weeks, spinal levels of interleukin-1β increased at both 4 weeks (acute phase) and 16 weeks (chronic phase). Importantly, systemic administration of anakinra, a peripherally restricted interleukin-1 receptor antagonist, only inhibited nociceptive behaviors at 4 weeks but not at 16 weeks, though intrathecal injection of anakinra reduced nociceptive behaviors at both 4 and 16 weeks.58  This study supports a critical role of central neuroinflammation in maintaining chronic pain in a rodent model of complex regional pain syndrome.

Emerging evidence suggests that neuroinflammation contributes to the pathophysiology of coprevalent or coexisting chronic pain conditions that are referred to as chronic overlapping pain conditions, which include but are not limited to fibromyalgia, headache, temporomandibular disorder, back pain, irritable bowel syndrome, primary headaches, pelvic pain, and vestibulodynia. Chronic overlapping pain conditions are characterized by symptoms consistent with the dysregulation of sensory, inflammatory, and psychologic domains.13,56,59,60  There is increased evidence that patients with chronic overlapping pain conditions exhibit increased basal and stress-induced levels of catecholamines (epinephrine and norepinephrine) in circulation61–64  and reduced activity of catechol-O-methyltransferase,65,66  a ubiquitously expressed enzyme that metabolizes catecholamines.67  Chronic overlapping pain condition patients have functional variants in the COMT gene that result in reduced catechol-O-methyltransferase activity.66, 68–70  The “low catechol-O-methyltransferase activity” variants are associated with increased fibromyalgia71–75  and temporomandibular disorder76  onset and increased pain in response to experimental stimuli76,77  and stressful events.70,78,79  Consistent with clinical syndromes, in rodents sustained delivery of the catechol-O-methyltransferase inhibitor OR486 results in pain at multiple body sites that persists for weeks and altered pain- and anxiety-related volitional behaviors.80,81–83  Persistent catechol-O-methyltransferase–dependent pain is initiated by peripheral β2- and β3-adrenergic receptors through release of nitric oxide, tumor necrosis factor-α, interleukin-1β, interleukin-6, and chemokine (CC motif) ligand 2 in plasma and maintained by increased tumor necrosis factor in central tissues.80,83–84  Higher levels of nitric oxide derivatives (e.g., nitrite and nitrate) and proinflammatory cytokines have been found in patients with chronic musculoskeletal pain conditions.13,85–89  Of note, patients with site-specific pain conditions (e.g., localized temporomandibular disorder or localized vestibulodynia) exhibit a balance in pro- and antiinflammatory cytokines, whereas those with chronic overlapping pain conditions fail to exhibit a compensatory increase in antiinflammatory cytokines.13,60  Compared to patients with localized pain, those with chronic overlapping pain conditions also exhibit dysregulation in microRNAs (e.g., miR-let-7f) that augment immune response and proinflammatory cytokine production.60  Together, these findings suggest that localized and anatomically widespread patterns of chronic pain are associated with distinct inflammatory profiles.

Widespread patterns of chronic pain exhibited by patients with chronic overlapping pain conditions may be attributed to previous injury or stressful events that produce long-lasting changes in nociceptor function, a phenomenon known as hyperalgesic priming. Preclinical studies have demonstrated that acute painful inflammation or nerve injury, as well as nonpainful stress, “primes” nociceptors so that they respond to a subsequent insult (e.g., prostaglandin E2 or epinephrine) for a prolonged duration due to activation of distinct kinase and gene transcription pathways.4,90–93  In line with these findings, individuals with the “low activity” catechol-O-methyltransferase genotype report enhanced and prolonged pain after motor vehicle collisions or psychologic strain,70,78,94  which are events that stimulate the sympathetic release of epinephrine known to sensitize nociceptors95,96  and promote inflammation.97–105  Thus, repeated environmental exposures to injury, inflammation, and stress in genetically predisposed individuals results in the transition from acute pain to chronic pain, in part through neuroinflammation (fig. 2). Of note, when a challenge of lipopolysaccharide is preceded by a surgical procedure, it has been shown that this early priming of spinal microglial cells increases the duration and intensity of pain through contributions to neuroinflammation.106 

Glial Activation, Gliosis, and Gliopathy after Painful Injuries

Neuroinflammation is characterized by activation of peripheral glia including Schwann cells in the nerve and satellite glial cells in the dorsal root ganglia and trigeminal ganglia, and central glia including microglia, astrocytes, and oligodendrocytes in the spinal cord and brain.107,108  In this review we focus on central glia, especially microglia and astrocytes and the mechanisms by which glia-produced mediators modulate synaptic plasticity and central sensitization. The previous decade has seen an exponential increase in literature documenting the role of microglia and astrocytes in the pathogenesis of chronic pain; “glial activation” is emerging as a powerful mechanism underlying pathogenesis of chronic pain,109–115  and chronic pain may also manifest as a “gliopathy.”108 

After painful injuries, there are different activation states of microglia and astrocytes. The morphologic activation (glial reaction or gliosis) is the most studied activation state, although this type of glial activation may not directly cause pain.108  Glial reaction is characterized by increased expression of microglial markers such as cluster of differentiation molecule 11B, ionized calcium-binder adapter molecule 1, and CX3C chemokine receptor 1 and astroglial markers glial fibrillary acid protein, as well as morphologic changes such as hypertrophy or process retraction/extension of microglia and astrocytes. Microglial reaction in the spinal cord is very rapid and dramatic, whereas astrocyte reaction in the spinal cord is more persistent and occurs in more painful conditions (fig. 3, A–F).114,116  Subcutaneous formalin injection into a hind paw of rat or mouse is probably the most studied animal model of inflammatory pain, which lasts for less than 1 h. Of interest, in 1999, Fu et al.117  showed that subcutaneous formalin also caused a robust microglial reaction (labeled with cluster of differentiation molecule 11B) in the spinal cord days after injection. This microglial reaction from formalin-induced nerve injury is associated with the development of mechanical allodynia. This is one of the earliest reports to support a possible role of spinal microglia in pain regulation. Functional imaging also reveals glial activation in patients with chronic pain.118  In 2003, Zhu et al.119  and Zhu and Eisenach120  showed that incision and nerve injury causes upregulation of cyclooxygenase-1 in spinal glial cells, which is important for the development of postoperative pain and neuropathic pain.

Fig. 3.

Distinct and time-dependent activation of microglia and astrocytes in the spinal cord after nerve injury. Microglia activation revealed by increased CX3C chemokine receptor 1 (CX3CR1) expression in the spinal cord 10 days (A) and 21 days (B) after nerve injury in Cx3cr1-GFP mice. Scale bar = 100 µm. (C) Phosphorylation of p38 mitogen-activated protein kinase (P-p38) in cluster of differentiation molecule 11B (CD11b)+ microglia in the spinal cord dorsal horn 7 days after nerve injury. Scale bar = 20 µm. Astroctye activation revealed by increased glial fibrillary acidic protein (GFAP) expression in the spinal cord 10 days (D) and 21 days (E) after nerve injury in mice. Scale bar = 100 µm. (F) Expression of connexin 43 (Cx43) in GFAP-positive astrocytes in the spinal cord dorsal horn 21 days after nerve injury. Scale bar = 20 µm. d = days.

Fig. 3.

Distinct and time-dependent activation of microglia and astrocytes in the spinal cord after nerve injury. Microglia activation revealed by increased CX3C chemokine receptor 1 (CX3CR1) expression in the spinal cord 10 days (A) and 21 days (B) after nerve injury in Cx3cr1-GFP mice. Scale bar = 100 µm. (C) Phosphorylation of p38 mitogen-activated protein kinase (P-p38) in cluster of differentiation molecule 11B (CD11b)+ microglia in the spinal cord dorsal horn 7 days after nerve injury. Scale bar = 20 µm. Astroctye activation revealed by increased glial fibrillary acidic protein (GFAP) expression in the spinal cord 10 days (D) and 21 days (E) after nerve injury in mice. Scale bar = 100 µm. (F) Expression of connexin 43 (Cx43) in GFAP-positive astrocytes in the spinal cord dorsal horn 21 days after nerve injury. Scale bar = 20 µm. d = days.

Close modal

Signaling Mechanisms in Glial Regulation of Allodynia and Hyperalgesia

Several neuromodulators such as adenosine triphosphate (ATP), chemokines (CL1, chemokine (CC motif) ligand 2, and chemokine (CXC motif) ligand 13), and neuropeptides (substance P and calcitonin gene–related peptide), and colony-stimulating factor 1 are involved in glial activation after painful insults108,121,122  (table 2). The upregulation of glia-specific receptors and channels are functionally correlated with pain hypersensitivity. ATP modulates glial activation via stimulation of ionotropic P2X receptors and metabotropic P2Y receptors.123,124  Peripheral nerve injury increases the expression of ATP P2Y receptors (P2X4, P2X7, and P2Y12) in spinal microglia, and each upregulation was implicated in neuropathic pain sensitization (mechanical allodynia).125,126  Accumulating evidence suggests that after tissue and nerve injury, ATP is generated from different cell types including astrocytes, neurons, and microglia. Astrocyte-expressing hemichannels connexin 43 are permeable to ATP.127  Glucocorticoids induce ATP release from spinal astrocytes, leading to microglial activation and diurnal exacerbation of allodynia.128  Vesicular nucleotide transporter regulates ATP release from spinal cord neurons after nerve injury.129  Microglial pannexin 1 channel was also shown to facilitate ATP release in neuropathic pain.130  Nerve injury results in cleavage and activation of chemokine CX3C ligand 1/fractalkine by protease cathepsin S, leading to microglia activation through stimulation of CX3C chemokine receptor 1 receptor.113  CX3C chemokine receptor 1, one of the best-known markers of microglia, is strongly upregulated after nerve injury, as revealed in Cx3cr1-GFP mice (fig. 3, A and B). This microglial receptor is also critical for the development of neuropathic pain symptoms, because mechanical allodynia after nerve injury is reduced after spinal administration of the CX3C chemokine receptor 1 neutralizing antibody and abrogated in Cx3cr1 knockout mice.113,131,132  After chronic constriction injury of the sciatic nerve, CX3C chemokine receptor 1 upregulation peaks within 10 days. In the late phase of mechanical allodynia (greater than 3 weeks), microglial CX3C chemokine receptor 1 expression markedly declines (fig. 3, A and B). Microglia may play a role in maintaining persistent hyperalgesia and allodynia after bone cancer.133  Orthopedic surgery and bone fracture also may result in nerve injury and microglial activation in the spinal cord. It was proposed that spinal microglial activation also contributes to postoperative cognitive dysfunction such as delirium.134  Substance P signaling from C-fiber afferent terminals in the spinal cord results in microglia activation and central sensitization after bone fracture.135 

Table 2.

Signal Transduction in Spinal Cord Microglia and Astrocytes after Tissue and Nerve Injury

Signal Transduction in Spinal Cord Microglia and Astrocytes after Tissue and Nerve Injury
Signal Transduction in Spinal Cord Microglia and Astrocytes after Tissue and Nerve Injury

After painful insults, gliopathy is also characterized by dysfunction of astrocytes, such as downregulation of glutamate transporters (glutamate transporter 1 and glutamate aspartate transporter) in spinal cord astrocytes, resulting in glutamate accumulation in synaptic clefts causing neuronal hyperactivity.108,136  Connexin 43 is a critical astrocytic signaling molecule that controls the release of astroglial mediators including glutamate and ATP.137  Of note, connexin 43 upregulation in spinal cord astrocytes is sustained after spinal cord injury and nerve injury (fig. 3F) and contributes to the development and maintenance of mechanical allodynia.138,139  Additionally, chronic pain-associated gliopathy could manifest as a functional switch of connexin 43 from gap junction communication to hemichannel regulation, so that astrocytes become “leaky” during this switch, resulting in increased secretion of cytokines (interleukin-1β) and chemokines ([CC motif] ligand 2, [CXC motif] ligand 1).139,140  Chemokines regulate bidirectional interactions of neurons and glial cells.141  Astrocytes not only produce chemokines that can “talk to” neurons by modulating neuronal activity but also “listen to” neurons by responding to chemokines (e.g., [CXC motif] ligand 13) derived from neurons.122  Astrocytes also release thrombospondin 4 to modulate synapse formation, synaptic plasticity, and behavioral hypersensitivity (table 2).142 

A critical step of glial activation in persistent pain is the activation of intracellular signaling pathways, especially the mitogen-activated protein kinase pathways. There are three major members in the mitogen-activated protein kinase family: extracellular signal-regulated kinase 1 and 2, p38, and c-Jun N-terminal kinase.143  Phosphorylation of p38 in spinal microglia occurs in different pain conditions after surgery, nerve injury (fig. 3C), and opioid tolerance, resulting in increased synthesis and release of microglial mediators (tumor necrosis factor, interleukin-1β, and brain-derived growth factor) and pain hypersensitivity.132,133,144–147  Inflammation or nerve injury also activates c-Jun N-terminal kinase in astrocytes,148,149  leading to increased secretion of chemokine (CC motif) ligand 2 and chemokine (CXC motif) ligand 1 and enhanced pain states.139,150  Nerve injury also causes sequential activation of extracellular signal-regulated kinase in microglia (early phase) and astrocytes (late phase),143  indicating distinct involvement of these two glial cell types in chronic pain induction and maintenance.

Glia Activation in the Brain after Painful Injuries

Accumulating evidence also suggests a role of glial activation, revealed in different brain regions, in regulating neuroinflammation and pain sensitivity. Sciatic nerve ligation induces astrocyte activation in the S1 sensory cortex, which is associated with upregulation of metabotropic glutamate receptor 5. Activation of this glutamate receptor subtype in astroglia induces spontaneous somatic Ca2+ transients and secretion of thrombospondin 1 from astrocytes, leading to new synapse formation and mechanical allodynia.151  Toll-like receptor 4 plays a critical role in glial activation and neuroinflammation in the spinal cord, as well as hyperalgesia and allodynia.152,153  Toll-like receptor 4 also contributes to neuroinflammation in the prefrontal cortex and visceral pain after chronic stress. Increased expression of Toll-like receptor 4 is associated enhanced glia activation in the prefrontal cortex and increased levels of proinflammatory cytokines. Administration of the Toll-like receptor 4–specific antagonist TAK-242 in the prefrontal cortex is sufficient to attenuate visceral hypersensitivity.154  Peripheral nerve injury causes microglia activation within the mesolimbic reward circuitry, leading to a disruption of dopaminergic signaling and reward behavior.155  Furthermore, nerve injury causes activation of microglia and astrocytes in the anterior cingulate cortex, and administration of microglial inhibitor minocycline in this brain region inhibited mechanical allodynia.151  However, earlier studies showed that nerve injury did not cause microgliosis in the anterior cingulate cortex and that long-term synaptic plasticity (long-term potentiation) was not altered by minocycline.156,157  Future studies are warranted to investigate how microglia and astrocytes regulate different forms of synaptic plasticity in different brain regions after painful insults in the peripheral tissues and the central nervous system.

Sex Dimorphism in Glial Regulation of Allodynia and Hyperalgesia

Chronic pain such as chronic orofacial pain associated with temporomandibular disorder occurs more frequently in women.158,159  In 1993, Maixner and Humphrey160  reported sex differences in pain and cardiovascular responses to forearm ischemia in humans. Paradoxically, the majority, if not all, pain-related studies were conducted in male animals.161  However, it was fortunate that males were tested because spinal microglia play little or no role in regulating inflammatory and neuropathic pain primarily in female rodents162–165  (fig. 4). Sorge162  demonstrated that spinal Toll-like receptor 4, an important receptor for microglia activation, regulates hyperalgesia and allodynia resulting from inflammation or nerve injury exclusively in male mice. Of note, morphologic activation and proliferation of spinal microglia are identical in males and females after nerve injury. Nerve injury–evoked mechanical allodynia is also equivalent in both sexes during the tested times.163,164  However, mechanical allodynia after nerve injury was exclusively attenuated in male mice after intrathecal injection of microglial inhibitor (minocycline), microglial toxin, or P2X4 blocker, or after special deletion of Bdnf in microglia.163  Furthermore, nerve injury activates p38 in spinal microglia of male but not female mice; in agreement, spinal administration of p38 inhibitor reduces neuropathic pain only in male mice.164  Caspase-6 is a microglial activator and released from axonal terminals in the spinal cord after tissue and nerve injury, which can act on microglia to release tumor necrosis factor.166  Sex dimorphism was also revealed in caspase-6–mediated microglial signaling, and caspase-6 regulates neuropathic pain exclusively in males.165  It appears some male-specific microglial responses require testosterone, because minocycline reduces allodynia in testosterone-treated females but not in castrated males. In female rodents, the role of microglia in neuropathic pain appears to be replaced by T cells.163 

Fig. 4.

Schematic illustration of local and remote central sensitization induced by glial activation and neuroinflammation in the spinal cord. Activation of spinal microglia and astrocytes by painful insults results in secretion of glial mediators such as tumor necrosis factor (TNF), interleukin (IL)-1β, chemokine (CC motif) ligand 2 (CCL2), chemokine (CXC motif) ligand 1 (CXCL1), brain-derived neurotrophic factor (BDNF), and d-serine, which can act as neuromodulators to induce local central sensitization in surrounding excitatory synapses (facilitation) and inhibitory synapses (disinhibition). During neuroinflammation, these glial mediators also affect synapses in different spinal segments to cause remote central sensitization and extraterritorial and widespread pain beyond the initial injury site. It is also possible that central sensitization may further promote peripheral sensitization via neuroinflammation. AMPAR = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; CSF = cerebrospinal fluid; GABA = γ-aminobutyric acid; GABAR = γ-aminobutyric acid receptor; Glu = glutamate; GlyR = glycine receptor; NMDAR = N-methyl-d-aspartate receptor.

Fig. 4.

Schematic illustration of local and remote central sensitization induced by glial activation and neuroinflammation in the spinal cord. Activation of spinal microglia and astrocytes by painful insults results in secretion of glial mediators such as tumor necrosis factor (TNF), interleukin (IL)-1β, chemokine (CC motif) ligand 2 (CCL2), chemokine (CXC motif) ligand 1 (CXCL1), brain-derived neurotrophic factor (BDNF), and d-serine, which can act as neuromodulators to induce local central sensitization in surrounding excitatory synapses (facilitation) and inhibitory synapses (disinhibition). During neuroinflammation, these glial mediators also affect synapses in different spinal segments to cause remote central sensitization and extraterritorial and widespread pain beyond the initial injury site. It is also possible that central sensitization may further promote peripheral sensitization via neuroinflammation. AMPAR = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; CSF = cerebrospinal fluid; GABA = γ-aminobutyric acid; GABAR = γ-aminobutyric acid receptor; Glu = glutamate; GlyR = glycine receptor; NMDAR = N-methyl-d-aspartate receptor.

Close modal

Term Development

Central sensitization is a powerful phenomenon in the pain field. As a key mechanism of chronic pain, it also guides clinical treatment for conditions associated with widespread pain.167,168  In this review, we highlight the role of glial cells and neuroinflammation in promoting central sensitization and widespread chronic pain. In 1983, Woolf 36  presented evidence for a central component of postinjury pain hypersensitivity. The International Association for the Study of Pain describes central sensitization as increased responsiveness of nociceptive neurons in the central nervous system to their normal or subthreshold afferent input.169  Central sensitization may also include conditions like increased central responsiveness due to dysfunction of endogenous pain control systems, regardless of whether there is functional change of peripheral neurons. In 2003, Ji et al.6  defined central sensitization as the increased synaptic efficacy established in somatosensory neurons in the dorsal horn of the spinal cord after intense peripheral noxious stimuli, tissue injury, or nerve damage. This heightened synaptic transmission results in a reduction in pain threshold, an amplification of pain responses, and a spread of pain sensitivity to noninjured areas. In 2009, Latremoliere and Woolf 38  described central sensitization as an enhancement in the function of neurons and circuits in nociceptive pathways caused by increases in membrane excitability and synaptic efficacy, as well as to reduced inhibition, and as a manifestation of the remarkable plasticity of the somatosensory nervous system in response to activity, inflammation, and neural injury. In this review, the authors highlighted disinhibition (reduced inhibition) and also emphasized that the net effect of central sensitization is to recruit previously subthreshold synaptic inputs to nociceptive neurons, generating an increased or augmented action potential output: a state of facilitation, potentiation, augmentation, or amplification.

Mechanisms of Central Sensitization

As shown in figure 5, activation of NMDA receptors is an essential step in initiating and maintaining the central sensitization and pain hypersensitivity after tissue and nerve injury.170,171  Glutamate is a primary excitatory neurotransmitter in the pain pathway. Under normal circumstances NMDA receptor channels are blocked by Mg2+ ions, but this blockade is removed by membrane depolarization after activation of nociceptive primary afferents. Activation of NMDA receptors boosts synaptic efficacy and causes Ca2+ influx, which can activate intracellular signaling pathways that initiate and maintain central sensitization.6,38  In particular, tissue and nerve injury increases the expression of the NMDA receptor-NR2B (GluN2B) subunit, which regulates spinal synaptic plasticity in persistent pain conditions together with the NR1 subunit.172  Interestingly, NR2B/GluN2B receptor activity and surface expression in spinal cord dorsal horn neurons is negatively regulated by β-arrestin 2,173  a scaffold protein that was traditionally known as an inhibitor of G-protein–coupled receptors. Deficiency of β-arrestin 2 results in enhanced acute opioid analgesia, produced by morphine and [D-Ala2, N-MePhe4, Gly-ol]-enkephalin, a selective µ opioid receptor agonist.173,174  Paradoxically, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin-induced hyperalgesia is also potentiated after β-arrestin 2 deficiency, as a result of enhanced surface and synapse expression of GluN2B that results in hyperactivity of the receptor. Loss of β-arrestin 2 also leads to a prolongation of inflammatory and neuropathic pain, because these pain conditions critically depend on GluN2B.173  It appears that for the resolution of persistent pain, it is more important for β-arrestin 2 to regulate NMDA receptors via extracellular signal-regulated kinase signaling pathway (fig. 5). Furthermore, surface trafficking of AMPA receptors, especially calcium-permeable subunit (GluR1/GluR-A), plays a critical role in spinal cord synaptic plasticity and pain hypersensitivity after tissue injury.175,176  The scaffold protein Homer1a operates in a negative feedback loop to regulate calcium signaling and the excitability of the spinal cord pain pathway in an activity-dependent manner.177  Given a critical role of AMPA receptor in direct control of excitatory synaptic transmission in pain circuits, NMDA receptor–independent central sensitization may also exist.

Fig. 5.

Molecular mechanisms of central sensitization in first-order excitatory synapses in the spinal cord dorsal horn pain circuit and induction of central sensitization by proinflammatory cytokines and chemokines (e.g., tumor necrosis factor [TNF], interleukin [IL]-1β, chemokine [CC motif] ligand 2 [CCL2], chemokine [CXC motif] ligand 1 [CXCL1]) that are produced by glial cells. At presynaptic sites (i.e., central terminals of nociceptive primary afferents), activation of receptors of cytokine and chemokine receptors results in phosphorylation and activation of extracellular signal-regulated kinase (P-ERK) and p38 (P-p38), leading to glutamate (Glu) release from synaptic vesicles via activation of ion channels transient receptor potential ion channel V1, voltage-gated sodium ion channel 1.7 (Nav1.7), and voltage-gated sodium channel subtype 1.8 (Nav1.8). At postsynaptic sites, increased release of neurotransmitters (e.g., glutamate) also induces phosphorylated extracellular signal-regulated kinase, which can induce central sensitization by positive modulation of N-methyl-d-aspartate receptor (NMDAR, step 1). Positive regulation of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR, step 2) and negative modulation of potassium channel subunit Kv4.2 (step 3) are also shown. Phosphorylated extracellular signal-regulated kinase also maintains central sensitization via inducing cAMP response element–binding protein phosphorylation (P-CREB, step 4). CREB is a critical transcription factor that controls the expression of pronociceptive genes. Opioids such as morphine inhibit neurotransmitter release via µ opioid receptors (MOR) and N-type calcium channels. The scaffold protein β-arrestin 2 (βarr2) inhibits µ opioid receptor signaling by desensitization and degradation of G-protein–coupled receptors, leading to enhanced acute opioid analgesia in β-arrestin 2 knockout mice. Paradoxically, β-arrestin 2 also inhibits N-methyl-d-aspartate receptor and extracellular signal-regulated kinase signaling, leading to a transition from acute pain to chronic pain.174  CXCR2 = CXC motif chemokine receptor 2; IL-1R = interleukin-1 receptor; TNFR = tumor necrosis factor receptor.

Fig. 5.

Molecular mechanisms of central sensitization in first-order excitatory synapses in the spinal cord dorsal horn pain circuit and induction of central sensitization by proinflammatory cytokines and chemokines (e.g., tumor necrosis factor [TNF], interleukin [IL]-1β, chemokine [CC motif] ligand 2 [CCL2], chemokine [CXC motif] ligand 1 [CXCL1]) that are produced by glial cells. At presynaptic sites (i.e., central terminals of nociceptive primary afferents), activation of receptors of cytokine and chemokine receptors results in phosphorylation and activation of extracellular signal-regulated kinase (P-ERK) and p38 (P-p38), leading to glutamate (Glu) release from synaptic vesicles via activation of ion channels transient receptor potential ion channel V1, voltage-gated sodium ion channel 1.7 (Nav1.7), and voltage-gated sodium channel subtype 1.8 (Nav1.8). At postsynaptic sites, increased release of neurotransmitters (e.g., glutamate) also induces phosphorylated extracellular signal-regulated kinase, which can induce central sensitization by positive modulation of N-methyl-d-aspartate receptor (NMDAR, step 1). Positive regulation of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR, step 2) and negative modulation of potassium channel subunit Kv4.2 (step 3) are also shown. Phosphorylated extracellular signal-regulated kinase also maintains central sensitization via inducing cAMP response element–binding protein phosphorylation (P-CREB, step 4). CREB is a critical transcription factor that controls the expression of pronociceptive genes. Opioids such as morphine inhibit neurotransmitter release via µ opioid receptors (MOR) and N-type calcium channels. The scaffold protein β-arrestin 2 (βarr2) inhibits µ opioid receptor signaling by desensitization and degradation of G-protein–coupled receptors, leading to enhanced acute opioid analgesia in β-arrestin 2 knockout mice. Paradoxically, β-arrestin 2 also inhibits N-methyl-d-aspartate receptor and extracellular signal-regulated kinase signaling, leading to a transition from acute pain to chronic pain.174  CXCR2 = CXC motif chemokine receptor 2; IL-1R = interleukin-1 receptor; TNFR = tumor necrosis factor receptor.

Close modal

Activation of intracellular pathways by protein kinases, such as protein kinase A, protein kinase C, Ca2+/calmodulin-dependent kinase II, Src (a tyrosine kinase encoded by sarcoma oncogene), and extracellular signal-regulated kinases (including extracellular signal-regulated kinases 1 and 2), is important for the generation of central sensitization.6,37,178  Notably, activation of extracellular signal-regulated kinase via phosphorylation of extracellular signal-regulated kinase in spinal cord dorsal horn neurons is nociceptive-specific and serves as a marker of central sensitization.179–181  Phosphorylation of extracellular signal-regulated kinase is a common pathway after the activation of various ionotropic and metabotropic receptors and protein kinases (protein kinase A, protein kinase C, and Src) as well as a downstream event of Ca2+ signaling.182–184  Phosphorylation of extracellular signal-regulated kinase induces central sensitization via rapid posttranslational regulation, such as suppression of potassium channel Kv4.2 activity, leading to hyperactivity of the spinal cord dorsal horn.185,186  Phosphorylation of extracellular signal-regulated kinase also contributes to rapid upregulation of NMDA receptor (GluN2B) function in spinal cord dorsal horn neurons in response to inflammatory mediators.150,187  Furthermore, translocation of phosphorylated extracellular signal-regulated kinase to the nuclei of spinal cord dorsal horn neurons activates the transcription factor cAMP response element–binding protein, leading to increased expression of pronociceptive genes encoding for c-Fos, NK-1, and prodynorphin.182,188 

Does Central Sensitization Require Peripheral Input?

Historically, it has been believed that the central sensitization to noxious stimuli requires sustained, intense, and repeated applications of the stimulus. More recently, it has become apparent that persistent peripheral nociceptive input may not be required to elicit central sensitization, because central sensitization can result from changes in the properties of neurons in the central nervous system that appear to be independent of peripheral input.38  Central sensitization produces pain hypersensitivity by changing the sensory responses elicited by normal inputs, including subthreshold innocuous tactile stimulation. For example, spinal cord disinhibition by intrathecal injection of γ-aminobutyric acid (GABA) and glycine receptor antagonists is sufficient to induce central sensitization and mechanical allodynia via disinhibition of inhibitory signaling and subsequent activation of excitatory signaling that is mediated by the NMDA receptor.189  Disinhibition of γ-aminobutyric acid–mediated (GABAergic) and glycinergic synaptic transmission in the spinal cord pain circuitry is critical to the generation of chronic pain.84–87  Gate control theory describes a tonic inhibition of the spinal cord pain circuit via inhibitory neurons.190  A feed-forward spinal cord glycinergic neural circuit in the laminae II–III dorsal horn gates mechanical allodynia, and nerve injury impairs glycinergic synaptic transmission and opens the gate to elicit mechanical allodynia.84 

It is noteworthy that some central etiologies/injuries such as spinal cord injury, traumatic brain injury, and multiple sclerosis may cause neuroinflammation, central sensitization, and chronic pain without a peripheral insult (fig. 2).191  Therefore, it is important for future studies to examine neuroinflammation in the brain, including those regions that do not receive input from primary afferents. Spinal cord injury is sufficient to produce hyperexcitability in primary sensory neurons192 via possible retrograde signaling from the dorsal root reflex. Neuroinflammation in the spinal cord may also regulate gene expression of primary sensory neurons via diffusible inflammatory mediators that can reach to dorsal root ganglia. Thus, there could be bidirectional interactions between peripheral sensitization and central sensitization. Central sensitization is not only secondary to peripheral sensitization but may, in turn, regulate peripheral sensitization (fig. 4).

An important step forward in revealing the role of central sensitization in widespread chronic pain is to demonstrate direct involvement of cytokines and chemokines (small cytokines) in the induction and maintenance of central sensitization.150,193–195  In 2001, Samad et al.193  showed that spinal interleukin-1β contributes to central sensitization and inflammatory pain hypersensitivity via transcriptional regulation that causes upregulations of cyclooxygenase-2 and prostaglandin E2. Notably, unilateral inflammation in the hind paw of rats caused widespread and bilateral increase of cyclooxygenase-2 in the spinal cord and brain, due to possible increase of interleukin-1β level in the cerebrospinal fluid.193  This may partially explain widespread pain in some chronic pain conditions (fig. 4). However, contribution of the spinal cyclooxygenase/prostaglandin E2 pathway to central sensitization is not supported by a clinical trial in postoperative pain.196  Despite a critical contribution of prostaglandin E2 to peripheral sensitization, the involvement of this important inflammatory mediator in regulating central sensitization is not well studied, but see the work of Ahmadi et al.197  In 2008, Kawasaki et al.194  demonstrated a direct induction of central sensitization by the proinflammatory cytokines tumor necrosis factor, interleukin-1β, and interleukin-6. These cytokines elicit very rapid increases (within 1 min) in excitatory synaptic transmission on spinal cord neurons. In support of this direct modulation, tumor necrosis factor, interleukin-1β, and interleukin-6 rapidly modulate the function of neurotransmitter receptors such as AMPA receptor, NMDA receptor, glycine receptor, and GABAR, which results in enhanced excitatory synaptic transmission and suppressed inhibitory synaptic transmission in the spinal pain circuit.194  In agreement with this finding, intrathecal injection of tumor necrosis factor, interleukin-1β, or interleukin-6 elicits rapid pain hypersensitivity in naive animals.194  Because these cytokines are elevated and circulating cerebrospinal fluid in chronic pain conditions,198,199  they are possible mediators of widespread pain, as a result of widespread central sensitization (fig. 4).

Tumor necrosis factor can be produced by microglia, astrocytes, and even dorsal root ganglia primary sensory neurons.200,201  However, in the spinal cord, tumor necrosis factor is primarily produced by microglia, as indicated by single-cell analysis of microglia, astrocytes, and neurons.166  Electrophysiologic analysis reveals that tumor necrosis factor increases glutamate release in transient receptor potential ion channel V1+ C-fiber terminals, leading to enhanced excitatory synaptic transmission in lamina IIo excitatory spinal cord dorsal horn interneurons.202  These lamina IIo interneurons synapse to lamina I projection neurons to form a pain circuit and potentiate pathologic pain.84  Tumor necrosis factor also increases NMDA currents in IIo excitatory interneurons via extracellular signal-regulated kinase activation.187  Additionally, tumor necrosis factor inhibits spontaneous action potentials in GABAergic neurons in the dorsal horn.203  Both type I and type II receptors of tumor necrosis factor are involved in behavioral manifestations of central sensitization after intrathecal tumor necrosis factor treatment or during formalin-induced second-phase pain.204  Notably, caspase-6 triggers tumor necrosis factor release from microglia to elicit central sensitization via tumor necrosis factor receptor signaling.166 

Interleukin-1β is expressed by both microglia and astrocytes in the spinal cord.110,147,205  Interleukin-18, a highly related family member of interleukin-1β, is induced in microglia by nerve injury and bone cancer.133,206  Caspase-1 cleaves and activates interleukin-1β and interleukin-18 and acts as a key component of inflammasome, which contributes to the pathogenesis of chronic pain.207  In addition, matrix metalloproteases 9 and 2 have been implicated in interleukin-1β cleavage and activation and regulation of glial activation in early versus late phase of neuropathic pain.208  Interleukin-1β induces central sensitization via both presynaptic modulation (increasing glutamate release)194  and postsynaptic regulation (phosphorylation of NMDA receptor209,210  and enhancement of NMDA current194 ). Endogenous interleukin-1β also potentiates presynaptic NMDA receptor function in neuropathic pain.211  Furthermore, interleukin-1β suppresses inhibitory synaptic transmission and GABA and glycine-induced currents in spinal lamina IIo neurons.194  Interleukin-18 also causes hyperactivity of spinal wide dynamic range neurons after mechanical stimuli in vivo.133  Thus, cytokines regulate central sensitization through multiple mechanisms that involve presynaptic and postsynaptic modulation as well as excitatory and inhibitory synaptic transmission modulation.

Astrocyte-produced chemokines (CC motif) ligand 2 and chemokine (CXC motif) ligand 1 also play an important role in central sensitization and chronic pain.139,150,212  Chemokine (CC motif) receptor 2, the major receptor for chemokine (CC motif) ligand 2 is expressed in neurons including dorsal root ganglia and spinal cord dorsal horn neurons.150,213,214  Bath application of chemokine (CC motif) ligand 2 to spinal cord slices induces rapid extracellular signal-regulated kinase activation and causes extracellular signal-regulated kinase-dependent potentiation of NMDA currents in dorsal horn neurons via the activation of chemokine (CC motif) receptor 2 receptors.150,214  Intrathecal injection of chemokine (CXC motif) ligand 1 induced extracellular signal-regulated kinase and cAMP response element–binding protein activation in spinal neurons via the stimulation of CXCR2 receptors,212  which is epigenetically regulated after injury.215  Notably, late-phase neuropathic pain and synaptic plasticity (excitatory postsynaptic current increase) in the spinal cord pain circuit, at 3 weeks after nerve injury, is transiently reversed by the CXCR2 antagonist SB225002. These results suggest an active role of chemokine (CXC motif) ligand 1/CXC motif chemokine receptor 2 in the maintenance of central sensitization.139 

Glial cells also produce growth factors such as brain-derived growth factor and basic fibroblast growth factor to enhance central sensitization and chronic pain.216,217  Brain-derived growth factor release from central terminals of primary afferents elicits central sensitization via activation of extracellular signal-regulated kinase and potentiation of NMDA receptor.182,218–220  Brain-derived growth factor signaling in spinal microglia also facilitates central sensitization, neuropathic pain, and morphine tolerance.216,221  Exposure of spinal lamina I projection neurons to brain-derived growth factor results in a depolarizing shift in the anion reversal potential, causing disinhibition of GABAergic system, a key regulatory mechanism in chronic pain.216,221  Brain-derived growth factor also modulates excitatory synaptic transmission in spinal cord dorsal horn lamina I neurons via activation of protein kinase Fyn.178 

Spinal cord long-term potentiation is an important form of synaptic plasticity and a unique form of central sensitization in chronic pain.222,223  Spinal cord long-term potentiation of C-fiber–evoked field potentials is typically induced by high-frequency tetanic stimulation of the sciatic nerve.224  Spinal cord long-term potentiation is also induced by nerve injury and opioid withdrawal.225–227  There are striking similarities between spinal cord long-term potentiation and central sensitization, and both show the critical requirements of NMDA receptor and involvement of key signaling transduction pathways including the protein kinase C, extracellular signal-regulated kinase, and Src, as well as dependence of protein synthesis and gene transcription.38,179,228  However, maintenance of late-phase long-term potentiation (greater than 4 h) may require additional mechanisms.225  It remains to be determined whether there is persistent spinal long-term potentiation months after painful insults, due to technical difficulty in following long-term potentiation over time.

Several lines of evidence suggest an essential role of neuroinflammation in inducing and sustaining spinal cord long-term potentiation. First, spinal tumor necrosis factor is both sufficient and required for the induction of spinal cord long-term potentiation via both tumor necrosis factor receptors type I and type II.202,229  Caspase-6 also contributes to the induction and maintenance of spinal cord long-term potentiation via tumor necrosis factor signaling.166  Second, interleukin-1β triggers spinal cord long-term potentiation not only in excitatory synapses230  but also in glycinergic synapses on GABAergic neurons in spinal cord slices,231  serving as another example of cytokine-induced disinhibition. Third, activation of spinal microglial CX3C chemokine receptor 1 via chemokine CX3C ligand 1/fractalkine is sufficient to elicit spinal cord long-term potentiation.232  Finally, chemokine (CC motif) receptor 2 is required for the maintenance of spinal cord long-term potentiation.214 

Recently, Kronschläger et al.233  demonstrated a gliogenic spinal cord long-term potentiation that can spread widely in nociceptive pathways. A fundamental feature of long-term potentiation induction in the brain is the requirement for coincident pre- and postsynaptic activity, which is important to restrict long-term potentiation expression to activated synapses only (homosynaptic long-term potentiation) as well as to define the input specificity of long-term potentiation. Gliogenic spinal cord long-term potentiation can travel long distances via cerebrospinal fluid, because this long-term potentiation can be induced by glial activation and diffusible messengers, such as d-serine and tumor necrosis factor.233  Strikingly, transfer of spinal cerebrospinal fluid from a donor animal displaying long-term potentiation is able to induce long-term potentiation in a naïve receiver animal.233  Therefore, this diffusible spinal cord long-term potentiation affects susceptible synapses at remote sites. Collectively, gliogenic long-term potentiation, as well as other forms of diffusible and glial-mediated central sensitization, may underlie widespread pain in chronic pain patients, especially in those with overlapping chronic pain conditions (fig. 6).

Fig. 6.

Nonpharmacologic approaches that can control neuroinflammation and produce multiple beneficial effects including prevention and resolution of chronic pain, prevention of neurodegeneration, and repair of cognitive function deficits. DHA = docosahexaenoic acid; DRG = dorsal root ganglia; EA = electroacupuncture; EPA = eicosapentaenoic acid; SPM = specialized proresolution mediator; TENS = transcutaneous electrical nerve stimulation; TMS = transcranial magnetic stimulation.

Fig. 6.

Nonpharmacologic approaches that can control neuroinflammation and produce multiple beneficial effects including prevention and resolution of chronic pain, prevention of neurodegeneration, and repair of cognitive function deficits. DHA = docosahexaenoic acid; DRG = dorsal root ganglia; EA = electroacupuncture; EPA = eicosapentaenoic acid; SPM = specialized proresolution mediator; TENS = transcutaneous electrical nerve stimulation; TMS = transcranial magnetic stimulation.

Close modal

Synaptic plasticity and long-term potentiation have also been investigated in the brain, such as in the anterior cingulate cortex,234  amygdala,235  and hippocampus236  after tissue and nerve injury. Further investigation is warranted to define the role of neuroinflammation and glial activation in these pain-associated synaptic changes in the brain.

The current levels of enthusiasm and interest in the development of new treatments that specifically target neuroinflammation and glial activation may be tempered by the somewhat disappointing results of previous clinical trials. Although animal models, as reviewed in the previous sections, demonstrated promising results in the reduction of neuropathic pain, glial activation, and neuroinflammation when treated by the glial modulator propentofylline,237,238  the same compound failed to provide beneficial reductions in neuropathic pain when administered to patients suffering from postherpetic neuralgia.239  Despite its efficacy in reducing postoperative pain in animal models, intrathecal injection of cyclooxygenase inhibitor such as ketorolac did not improve acute or chronic pain after hip arthroplastry.119,196  Although the clinical trial does not support a central role of cyclooxygenase/prostaglandin E2 pathway in clinical pain, cytokines and chemokines can independently regulate central sensitization via direct actions on nociceptive neurons (fig. 5). Thus, future clinical studies are still needed to test the effects of spinal inhibition of cytokines/chemokines in clinical pain. Neuropathic pain patients demonstrate tolerance to p38 inhibitors including dilmapimod and losmapimod,240,241  but these drugs have inconsistent levels of efficacy. An exploratory trial of dilmapimod significantly inhibited nerve injury–induced neuropathic pain240 ; however, losmapimod did not demonstrate any significant analgesic effect in comparison to placebo controls.241  Intriguingly, acute postsurgical dental pain was significantly reduced when treated with another p38 inhibitor, specifically the novel p38α mitogen-activated protein kinase inhibitor SCIO-469.242 

The use of cytokine inhibitors and glial modulators in clinical trials has shown some encouraging results. Intractable discogenic lower back pain patients showed up to 8 weeks of pain relief when given a single intradiscal treatment of entanercept, a tumor necrosis factor inhibitor.243  Additionally, lumbar disc herniation patients who received entanercept via transforaminal epidural injections had up to 26 weeks of pain relief after two injections in a randomized, double-blind, and placebo-controlled trial.244  Another cytokine inhibitor, the interleukin-1 trap rilonacept, is well tolerated by patients, and in a proof-of-concept study, treatment with the drug showed pain relief for a small group of patients being treated for chronic refractory gouty arthritis.245  By contrast, subcutaneous inhibition of interleukin-1β with anakinra has no beneficial effect on chronic fatigue syndrome.246  Microglial activation can be blocked in vitro by low doses of naltrexone,247  and a pilot trial also showed that treatment with the drug can help to reduce symptoms related to fibromyalgia.248 

Certain limitations and concerns regarding the design of the mentioned trials need to be addressed. The first concern regards the lack of neuroinflammation analysis by biomarkers, because inhibition of in vivo glial responses in the propentyofylline study notably had no biomarker validation.239  A second concern is the lack of validation regarding whether central sites, including the spinal cord, received exposure to systemically administered drugs. For instance, it was noted by the authors of the losmapimod study that the lack of response may be a result of central sites having a lack of adequate exposure to the drug.241  In the majority of preclinical studies, inhibitors of glial cells, mitogen-activated protein kinases, cytokines, and chemokines are given via intrathecal administration, which ensures direct exposure of central sites to the drug being studied.108  The third concern is the absence of analysis of sex-dependent effects of treatments in previous studies of p38 and glial inhibitors in light of emerging evidence of sex dimorphism in microglial and p38 signaling and T-cell signaling in inflammatory and neuropathic pain.163,164  Fourth, the timing of administration appears to be a critical component of a treatment’s efficacy. During the acute phase of pain, inhibitors of microglia, p38 mitogen-activated protein kinase, and tumor necrosis factor-α show stronger efficacy, with more partial effects in the chronic phase according to preclinical studies.108,249  In line with this point, the effective results of the p38 inhibitor SCIO-46 trial were demonstrated in dental patients during the acute phase of postsurgical pain.242  It should be noted, however, that the intrathecal administration of the interleukin-1 receptor antagonist anakinra at 16 weeks post–bone fracture in rodents can reduce chronic postoperative pain.58  Finally, the mechanisms of the tested drugs need to be further elucidated. Propentofylline, although a glial modulator, also acts as a phopsphodiesterase inhibitor, as well as an adenosine uptake inhibitor. Thus, the administration of this drug may alter cAMP and adenosine levels in both glial and nonglial cells.250 

Animal models are of exceeding importance in the understanding of the mechanisms of pain, as well as for testing newly developed therapeutics. However, animal models cannot reproduce all key clinical symptoms of pain, and thus the gap in translation between preclinical studies in rodents and clinical studies in humans is a major topic in pain research discussed in numerous review articles.251,252  With regard to methods of measurement, in animal studies pain is measured by application of von Frey filaments to cause reflex pain with subsequent quantification of paw withdrawal thresholds, whereas most clinical studies measure pain based on the visual pain scale. Additionally, the time course of pain is very different between animals and humans, with the duration of pain in preclinical studies lasting normally from days to weeks, whereas clinical patients with chronic pain often suffer from their symptoms from months to years. Thus, developing animal models that accurately reflect the various genetic, sex-dependent, psychologic, and environmental factors and sequelae of the development and maintenance of chronic pain proves to pose a continuing challenge. Although clinically effective treatments do show efficacy in animal models thus far, increasing efforts to increase the translatability of preclinical and clinical studies will prove important, for instance measurement of spontaneous pain in animal models and quantitative sensory testing in human patients.59,253–256 

Although direct targeting of neuroinflammation via inhibitors of cytokines, chemokines, and mitogen-activated protein kinases could be effective, these drugs may also produce side effects such as infection after long-term treatment and impair the resolution of inflammation.39  In this review, we highlight the alternative approaches that can control excessive neuroinflammation, including specialized proresolution mediators, cell therapies, and neuromodulation (fig. 6).

Specialized proresolution mediators, including lipoxins, resolvins, protectins, and maresins, are biosynthesized from polyunsaturated fatty acids, including the omega-3 fatty acids docosahexaenoic acid and eicosapentaenoic acid, two major components of fish oil. Specialized proresolution mediators have multiple beneficial actions for treating inflammation-associated disease conditions.257,258  Lipoxin A4, resolvin D1, resolvin E1, resolvin D2, neuroprotectin D1, and maresin 1, at very low doses (10 to 500, ng range), reduced inflammatory pain, postoperative pain, and neuropathic pain in animal models.187,202,258–263  In the hippocampus, treatment with resolvin D1 prevented long-term potentiation impairment by reducing proinflammatory cytokines after surgery.264  The benefits of these specialized proresolution mediators include high potency, favorable safety profile, and multiple mechanisms of action including but not limited to control of inflammation in peripheral tissues, control of neuroinflammation in the peripheral nervous system and CNS, resolution of synaptic plasticity, modulation of transient receptor potential ion channel A1 and V1 activities, and protection against nerve injury.39,226,258,264  Spinal administration of neuroprotectin D1, even 2 h after long-term potentiation induction, is also effective in reversing long-term potentiation in the intact spinal cord.202,261  Specialized proresolution mediators are especially effective in preventing neuroinflammation, postoperative pain, and neuropathic pain after nerve injury and surgery, although posttreatment of specialized proresolution mediators also exhibit transient analgesic effects. Notably, thoracotomies produce a high incidence of chronic postoperative pain by nerve compression.265  Wang and Strichartz263  found that intrathecal and perioperative treatment with resolvin D1 and resolvin D2 effectively prevents postoperative pain in a rat model of chronic postthoracotomy.

Cell therapies are emerging treatments for chronic pain and neurodegenerative conditions. Implantation of bone marrow stem cells produces long-term pain relief.266,267  Bone marrow stromal cells or bone marrow stem cells promote tissue regeneration and tissue repair by secreting growth factors and control inflammation and neuroinflammation by secreting antiinflammatory mediators such as transforming growth factor β1.266  A single intrathecal injection of bone marrow stem cells not only inhibits nerve injury-induced neuropathic pain for many weeks via secretion of transforming growth factor β1 but also blocks nerve injury-induced neuroinflammation (glial activation and cytokine/chemokine upregulation) in dorsal root ganglia and spinal cord tissues.266  This paracrine modulation of neuroinflammation by bone marrow stem cells is very different from typical cell replacement strategies, such as implantation of forebrain GABAergic precursor cells into the spinal cord to generate functional GABAergic neurons for chronic pain control.268  Furthermore, subcutaneous treatment with human umbilical cord blood–derived multipotent stem cells reduced peripheral neuropathic pain in rats and inhibited spinal matrix metalloprotease 9 and 2 expression after nerve injury.269  Macrophages and T cells also play a role in the resolution of pain. Proresolution macrophages (M2-like) may inhibit neuroinflammation and chronic pain by secreting specialized proresolution mediators and antiinflammatory cytokines such as interleukin-10.26,270  Adoptive transfer of regulatory T cells (Treg) reduced neuropathic pain, whereas CD8+ cytotoxic T cells increased neuropathic pain after chemotherapy after intrathecal injection.30  However, adoptive transfer of CD8+ T cells via systemic route was also shown to resolve chemotherapy-induced neuropathic pain by increasing interleukin-10 receptor expression in dorsal root ganglia neurons.271  Autologous conditioned serum is prepared from whole blood that is incubated with glass beads to initiate monocyte activation. Autologous conditioned serum contains increased levels of interleukin-1 receptor antagonist interleukin-1ra as well as the antiinflammatory cytokines interleukin-4 and interleukin-10 and demonstrates efficacy in relieving pain in patients with knee osteoarthritis.272,273  Platelet-rich plasma was also shown to reduce clinical pain in knee osteoarthritis via possible modulation of inflammatory responses.274 

Neuromodulation via electrical and magnetic stimulation, such as spinal cord stimulation, deep brain stimulation, transcranial magnetic stimulation, transcutaneous electrical nerve stimulation, vagus nerve stimulation, auricular stimulation, and dorsal root ganglia stimulation, as well as acupuncture including electroacupuncture, has been used to provide pain relief in patients and animals275,276 via activation of specific neural pathways,277,278  suppression of nociceptive neuron activities (e.g., wide dynamic neurons and projection neurons in the spinal cord279,280 ), and release of pain suppressing neurotransmitters and neuromodulators.281  However, transient modulation of neuronal activity in the pain circuits during stimulation cannot explain the long-term benefits of neuromodulation. Increasing evidence suggests that neuromodulation such as vagus nerve stimulation can powerfully regulate inflammation.282,283  Although acupuncture elicits transient analgesia via releasing opioid peptides and adenosine,281,284  acupuncture and sciatic nerve activation also regulates inflammation and sepsis through vagus nerve activation and dopamine release.285  In 1984, Maixner and Randich286  demonstrated an antinociceptive role of vagal stimulation. Notably, acupuncture also causes neuronal activation in the nucleus of solitary tract to mediate vagal responses.287,288  In a rodent model of inflammatory muscle pain, acupuncture elicits antiinflammatory effects via interleukin-10 release.289  The identification of the “inflammatory reflex” reveals a neural circuit capable of providing information in real time to the brain about the body’s inflammatory status, allowing rapid neural regulatory responses via the vagus nerve.290  This has clear implications in developing novel bioelectronic technologies to treat pain and improve postoperative outcomes in vulnerable patients.283 

Neuroinflammation resulting from neuroglial and neuroimmune interactions not only serves as a driving force for chronic pain but is also implicated in other neurologic and psychiatric diseases such as Alzheimer disease, Parkinson disease, multiple sclerosis, autism, major depression, and schizophrenia.39  Chronic pain is commonly associated with depression, anxiety, and sleep disorders, and the prevalence of chronic pain is especially high in the rapidly growing populations of aged people and females with overlapping chronic pain conditions. Thus, targeting excessive neuroinflammation will help to alleviate chronic pain and control the progression of neurologic and psychiatric diseases in aged as well as younger populations. Mechanistically, neuroinflammation drives widespread chronic pain via central sensitization, which can be induced and maintained by cytokines, chemokines, and other glia-produced mediators circulated in the cerebrospinal fluid. Therefore, increasing the precision with which drugs can target neuroinflammation in the CNS, namely by increasing access to the spinal cord and brain, and developing the ability to accurately measure evolving neuroinflammatory changes in the CNS particularly in the cerebrospinal fluid will be of great importance. By developing specific neuroinflammatory profiles, their creation may also reveal novel biomarkers and means to identify chronic pain states. For instance, a recent study utilizing functional imaging of patients with chronic low-back pain revealed glial activation in the brain.118  In line with the use of an analgesic marker of glial activation, translocator protein,291  the development of additional novel radioligands to characterize different activation states of glial cells in the brain, as well as in spinal cord, dorsal root ganglia, and peripheral nerves, is of pressing need. Specialized proresolution mediators including resolvins and protectins, as well as bone marrow stem cells and neuromodulation, can serve as alternative approaches to providing effective control of neuroinflammation with perhaps less side effects than more directly targeted neuroinflammation treatments including cytokine, chemokine, and mitogen-activated protein kinase inhibitors. With the high incidence of postoperative pain after trauma and nerve injury resulting from surgeries265  and the worsening opioid crisis in the United States,292  the development of effective nonopioid treatments for the prevention and resolution of neuroinflammation and postoperative pain is of the utmost importance for clinical practice and health care. The benefits of developing novel treatments may extend further toward improving cognitive function in postoperative patients, a topic that will be addressed by the authors in future reviews.

Supported in part by grant Nos. R01DE17794, R01DE22743, and R01NS87988 from the National Institutes of Health, Bethesda, Maryland (to Dr. Ji).

The authors declare no competing interests.

1.
Pizzo
PA
,
Clark
NM
:
Alleviating suffering 101: Pain relief in the United States.
N Engl J Med
2012
;
366
:
197
9
2.
Gereau
RWt
,
Sluka
KA
,
Maixner
W
,
Savage
SR
,
Price
TJ
,
Murinson
BB
,
Sullivan
MD
,
Fillingim
RB
:
A pain research agenda for the 21st century.
J Pain
2014
;
15
:
1203
14
3.
Basbaum
AI
,
Bautista
DM
,
Scherrer
G
,
Julius
D
:
Cellular and molecular mechanisms of pain.
Cell
2009
;
139
:
267
84
4.
Hucho
T
,
Levine
JD
:
Signaling pathways in sensitization: Toward a nociceptor cell biology.
Neuron
2007
;
55
:
365
76
5.
Gold
MS
,
Gebhart
GF
:
Nociceptor sensitization in pain pathogenesis.
Nat Med
2010
;
16
:
1248
57
6.
Ji
RR
,
Kohno
T
,
Moore
KA
,
Woolf
CJ
:
Central sensitization and LTP: Do pain and memory share similar mechanisms?
Trends Neurosci
2003
;
26
:
696
705
7.
Woolf
CJ
:
Central sensitization: Implications for the diagnosis and treatment of pain.
Pain
2011
;
152
(
suppl 3
):
S2
15
8.
Ossipov
MH
,
Dussor
GO
,
Porreca
F
:
Central modulation of pain.
J Clin Invest
2010
;
120
:
3779
87
9.
Luo
C
,
Kuner
T
,
Kuner
R
:
Synaptic plasticity in pathological pain.
Trends Neurosci
2014
;
37
:
343
55
10.
Oh
SB
,
Tran
PB
,
Gillard
SE
,
Hurley
RW
,
Hammond
DL
,
Miller
RJ
:
Chemokines and glycoprotein120 produce pain hypersensitivity by directly exciting primary nociceptive neurons.
J Neurosci
2001
;
21
:
5027
35
11.
Dawes
JM
,
Calvo
M
,
Perkins
JR
,
Paterson
KJ
,
Kiesewetter
H
,
Hobbs
C
,
Kaan
TK
,
Orengo
C
,
Bennett
DL
,
McMahon
SB
:
CXCL5 mediates UVB irradiation-induced pain.
Sci Transl Med
2011
;
3
:
90ra60
12.
Angst
MS
,
Clark
JD
,
Carvalho
B
,
Tingle
M
,
Schmelz
M
,
Yeomans
DC
:
Cytokine profile in human skin in response to experimental inflammation, noxious stimulation, and administration of a COX-inhibitor: A microdialysis study.
Pain
2008
;
139
:
15
27
13.
Slade
GD
,
Conrad
MS
,
Diatchenko
L
,
Rashid
NU
,
Zhong
S
,
Smith
S
,
Rhodes
J
,
Medvedev
A
,
Makarov
S
,
Maixner
W
,
Nackley
AG
:
Cytokine biomarkers and chronic pain: Association of genes, transcription, and circulating proteins with temporomandibular disorders and widespread palpation tenderness.
Pain
2011
;
152
:
2802
12
14.
Pan
HL
,
Liu
BL
,
Lin
W
,
Zhang
YQ
:
Modulation of Nav1.8 by lysophosphatidic acid in the induction of bone cancer pain.
Neurosci Bull
2016
;
32
:
445
54
15.
Kim
SE
,
Coste
B
,
Chadha
A
,
Cook
B
,
Patapoutian
A
:
The role of Drosophila Piezo in mechanical nociception.
Nature
2012
;
483
:
209
12
16.
Xu
XZ
:
Demystifying mechanosensitive Piezo ion channels.
Neurosci Bull
2016
;
32
:
307
9
17.
Park
CK
,
Xu
ZZ
,
Berta
T
,
Han
Q
,
Chen
G
,
Liu
XJ
,
Ji
RR
:
Extracellular microRNAs activate nociceptor neurons to elicit pain via TLR7 and TRPA1.
Neuron
2014
;
82
:
47
54
18.
Jin
X
,
Gereau
RW
IV
:
Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-α.
J Neurosci
2006
;
26
:
246
55
19.
Liu
XJ
,
Liu
T
,
Chen
G
,
Wang
B
,
Yu
XL
,
Yin
C
,
Ji
RR
:
TLR signaling adaptor protein MyD88 in primary sensory neurons contributes to persistent inflammatory and neuropathic pain and neuroinflammation.
Sci Rep
2016
;
6
:
28188
20.
Ji
RR
,
Samad
TA
,
Jin
SX
,
Schmoll
R
,
Woolf
CJ
:
p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia.
Neuron
2002
;
36
:
57
68
21.
Obata
K
,
Katsura
H
,
Mizushima
T
,
Yamanaka
H
,
Kobayashi
K
,
Dai
Y
,
Fukuoka
T
,
Tokunaga
A
,
Tominaga
M
,
Noguchi
K
:
TRPA1 induced in sensory neurons contributes to cold hyperalgesia after inflammation and nerve injury.
J Clin Invest
2005
;
115
:
2393
401
22.
Woolf
CJ
,
Allchorne
A
,
Safieh-Garabedian
B
,
Poole
S
:
Cytokines, nerve growth factor and inflammatory hyperalgesia: The contribution of tumour necrosis factor α.
Br J Pharmacol
1997
;
121
:
417
24
23.
Schäfers
M
,
Sorkin
LS
,
Sommer
C
:
Intramuscular injection of tumor necrosis factor-α induces muscle hyperalgesia in rats.
Pain
2003
;
104
:
579
88
24.
Schäfers
M
,
Svensson
CI
,
Sommer
C
,
Sorkin
LS
:
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
21
25.
Sommer
C
,
Schmidt
C
,
George
A
:
Hyperalgesia in experimental neuropathy is dependent on the TNF receptor 1.
Exp Neurol
1998
;
151
:
138
42
26.
Binshtok
AM
,
Wang
H
,
Zimmermann
K
,
Amaya
F
,
Vardeh
D
,
Shi
L
,
Brenner
GJ
,
Ji
RR
,
Bean
BP
,
Woolf
CJ
,
Samad
TA
:
Nociceptors are interleukin-1β sensors.
J Neurosci
2008
;
28
:
14062
73
27.
Ji
RR
,
Chamessian
A
,
Zhang
YQ
:
Pain regulation by non-neuronal cells and inflammation.
Science
2016
;
354
:
572
7
28.
Chiu
IM
,
von Hehn
CA
,
Woolf
CJ
:
Neurogenic inflammation and the peripheral nervous system in host defense and immunopathology.
Nat Neurosci
2012
;
15
:
1063
7
29.
Liu
T
,
Ji
RR
:
New insights into the mechanisms of itch: Are pain and itch controlled by distinct mechanisms?
Pflugers Arch
2013
;
465
:
1671
85
30.
Liu
T
,
Berta
T
,
Xu
ZZ
,
Park
CK
,
Zhang
L
,
N
,
Liu
Q
,
Liu
Y
,
Gao
YJ
,
Liu
YC
,
Ma
Q
,
Dong
X
,
Ji
RR
:
TLR3 deficiency impairs spinal cord synaptic transmission, central sensitization, and pruritus in mice.
J Clin Invest
2012
;
122
:
2195
207
31.
Liu
XJ
,
Zhang
Y
,
Liu
T
,
Xu
ZZ
,
Park
CK
,
Berta
T
,
Jiang
D
,
Ji
RR
:
Nociceptive neurons regulate innate and adaptive immunity and neuropathic pain through MyD88 adapter.
Cell Res
2014
;
24
:
1374
7
32.
Talbot
S
,
Foster
SL
,
Woolf
CJ
:
Neuroimmunity: Physiology and pathology.
Annu Rev Immunol
2016
;
34
:
421
47
33.
Zhang
H
,
Boyette-Davis
JA
,
Kosturakis
AK
,
Li
Y
,
Yoon
SY
,
Walters
ET
,
Dougherty
PM
:
Induction of monocyte chemoattractant protein-1 (MCP-1) and its receptor CCR2 in primary sensory neurons contributes to paclitaxel-induced peripheral neuropathy.
J Pain
2013
;
14
:
1031
44
34.
Wei
T
,
Li
WW
,
Guo
TZ
,
Zhao
R
,
Wang
L
,
Clark
DJ
,
Oaklander
AL
,
Schmelz
M
,
Kingery
WS
:
Post-junctional facilitation of Substance P signaling in a tibia fracture rat model of complex regional pain syndrome type I.
Pain
2009
;
144
:
278
86
35.
Chiu
IM
,
Heesters
BA
,
Ghasemlou
N
,
Von Hehn
CA
,
Zhao
F
,
Tran
J
,
Wainger
B
,
Strominger
A
,
Muralidharan
S
,
Horswill
AR
,
Bubeck Wardenburg
J
,
Hwang
SW
,
Carroll
MC
,
Woolf
CJ
:
Bacteria activate sensory neurons that modulate pain and inflammation.
Nature
2013
;
501
:
52
7
36.
Woolf
CJ
:
Evidence for a central component of post-injury pain hypersensitivity.
Nature
1983
;
306
:
686
8
37.
Woolf
CJ
,
Salter
MW
:
Neuronal plasticity: Increasing the gain in pain.
Science
2000
;
288
:
1765
9
38.
Latremoliere
A
,
Woolf
CJ
:
Central sensitization: A generator of pain hypersensitivity by central neural plasticity.
J Pain
2009
;
10
:
895
926
39.
Ji
RR
,
Xu
ZZ
,
Gao
YJ
:
Emerging targets in neuroinflammation-driven chronic pain.
Nat Rev Drug Discov
2014
;
13
:
533
48
40.
Vargas
DL
,
Nascimbene
C
,
Krishnan
C
,
Zimmerman
AW
,
Pardo
CA
:
Neuroglial activation and neuroinflammation in the brain of patients with autism.
Ann Neurol
2005
;
57
:
67
81
41.
Culley
DJ
,
Snayd
M
,
Baxter
MG
,
Xie
Z
,
Lee
IH
,
Rudolph
J
,
Inouye
SK
,
Marcantonio
ER
,
Crosby
G
:
Systemic inflammation impairs attention and cognitive flexibility but not associative learning in aged rats: Possible implications for delirium.
Front Aging Neurosci
2014
;
6
:
107
42.
Chiu
IM
:
Infection, pain, and itch.
Neurosci Bull
2018
;
34
:
109
19
43.
Watkins
LR
,
Hutchinson
MR
,
Rice
KC
,
Maier
SF
:
The “toll” of opioid-induced glial activation: Improving the clinical efficacy of opioids by targeting glia.
Trends Pharmacol Sci
2009
;
30
:
581
91
44.
Song
P
,
Zhao
ZQ
:
The involvement of glial cells in the development of morphine tolerance.
Neurosci Res
2001
;
39
:
281
6
45.
Sisignano
M
,
Baron
R
,
Scholich
K
,
Geisslinger
G
:
Mechanism-based treatment for chemotherapy-induced peripheral neuropathic pain.
Nat Rev Neurol
2014
;
10
:
694
707
46.
Huang
W
,
Calvo
M
,
Karu
K
,
Olausen
HR
,
Bathgate
G
,
Okuse
K
,
Bennett
DL
,
Rice
AS
:
A clinically relevant rodent model of the HIV antiretroviral drug stavudine induced painful peripheral neuropathy.
Pain
2013
;
154
:
560
75
47.
Sharma
P
,
Allison
JP
:
The future of immune checkpoint therapy.
Science
2015
;
348
:
56
61
48.
Patel
SP
,
Kurzrock
R
:
PD-L1 expression as a predictive biomarker in cancer immunotherapy.
Mol Cancer Ther
2015
;
14
:
847
56
49.
Uçeyler
N
,
Göbel
K
,
Meuth
SG
,
Ortler
S
,
Stoll
G
,
Sommer
C
,
Wiendl
H
,
Kleinschnitz
C
:
Deficiency of the negative immune regulator B7-H1 enhances inflammation and neuropathic pain after chronic constriction injury of mouse sciatic nerve.
Exp Neurol
2010
;
222
:
153
60
50.
Chen
GKY
,
Ji
RR
:
PD-L1 inhibits acute and chronic pain and suppresses nociceptor activity via PD-1.
Nat Neurosci
2017
;
20
:
917
26
51.
Rees
H
,
Sluka
KA
,
Westlund
KN
,
Willis
WD
:
Do dorsal root reflexes augment peripheral inflammation?
Neuroreport
1994
;
5
:
821
4
52.
Sluka
KA
,
Willis
WD
,
Westlund
KN
:
Central control of peripheral joint inflammation and heat hyperalgesia.
Prog Pain Res Manag
1994
;
2
:
359
71
53.
Xanthos
DN
,
Sandkühler
J
:
Neurogenic neuroinflammation: Inflammatory CNS reactions in response to neuronal activity.
Nat Rev Neurosci
2014
;
15
:
43
53
54.
Christianson
CA
,
Dumlao
DS
,
Stokes
JA
,
Dennis
EA
,
Svensson
CI
,
Corr
M
,
Yaksh
TL
:
Spinal TLR4 mediates the transition to a persistent mechanical hypersensitivity after the resolution of inflammation in serum-transferred arthritis.
Pain
2011
;
152
:
2881
91
55.
Shi
Y
,
Gelman
BB
,
Lisinicchia
JG
,
Tang
SJ
:
Chronic-pain-associated astrocytic reaction in the spinal cord dorsal horn of human immunodeficiency virus-infected patients.
J Neurosci
2012
;
32
:
10833
40
56.
Sluka
KA
,
Clauw
DJ
:
Neurobiology of fibromyalgia and chronic widespread pain.
Neuroscience
2016
;
338
:
114
29
57.
Üçeyler
N
,
Zeller
D
,
Kahn
AK
,
Kewenig
S
,
Kittel-Schneider
S
,
Schmid
A
,
Casanova-Molla
J
,
Reiners
K
,
Sommer
C
:
Small fibre pathology in patients with fibromyalgia syndrome.
Brain
2013
;
136
:
1857
67
58.
Wei
T
,
Guo
TZ
,
Li
WW
,
Kingery
WS
,
Clark
JD
:
Acute versus chronic phase mechanisms in a rat model of CRPS.
J Neuroinflammation
2016
;
13
:
14
59.
Maixner
W
,
Fillingim
RB
,
Williams
DA
,
Smith
SB
,
Slade
GD
:
Overlapping chronic pain conditions: Implications for diagnosis and classification.
J Pain
2016
;
17
(
suppl 9
):
T93
T107
60.
Ciszek
BP
,
Khan
AA
,
Dang
H
,
Slade
GD
,
Smith
S
,
Bair
E
,
Maixner
W
,
Zolnoun
D
,
Nackley
AG
:
MicroRNA expression profiles differentiate chronic pain condition subtypes.
Transl Res
2015
;
166
:
706
720.e11
61.
Torpy
DJ
,
Papanicolaou
DA
,
Lotsikas
AJ
,
Wilder
RL
,
Chrousos
GP
,
Pillemer
SR
:
Responses of the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis to interleukin-6: A pilot study in fibromyalgia.
Arthritis Rheum
2000
;
43
:
872
80
62.
Evaskus
DS
,
Laskin
DM
:
A biochemical measure of stress in patients with myofascial pain-dysfunction syndrome.
J Dent Res
1972
;
51
:
1464
6
63.
Perry
F
,
Heller
PH
,
Kamiya
J
,
Levine
JD
:
Altered autonomic function in patients with arthritis or with chronic myofascial pain.
Pain
1989
;
39
:
77
84
64.
Bote
ME
,
García
JJ
,
Hinchado
MD
,
Ortega
E
:
An exploratory study of the effect of regular aquatic exercise on the function of neutrophils from women with fibromyalgia: Role of IL-8 and noradrenaline.
Brain Behav Immun
2014
;
39
:
107
12
65.
Marbach
JJ
,
Levitt
M
:
Erythrocyte catechol-O-methyltransferase activity in facial pain patients.
J Dent Res
1976
;
55
:
711
66.
Smith
SB
,
Reenilä
I
,
Männistö
PT
,
Slade
GD
,
Maixner
W
,
Diatchenko
L
,
Nackley
AG
:
Epistasis between polymorphisms in COMT, ESR1, and GCH1 influences COMT enzyme activity and pain.
Pain
2014
;
155
:
2390
9
67.
Männistö
PT
,
Kaakkola
S
:
Catechol-O-methyltransferase (COMT): Biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors.
Pharmacol Rev
1999
;
51
:
593
628
68.
Lotta
T
,
Vidgren
J
,
Tilgmann
C
,
Ulmanen
I
,
Melén
K
,
Julkunen
I
,
Taskinen
J
:
Kinetics of human soluble and membrane-bound catechol O-methyltransferase: A revised mechanism and description of the thermolabile variant of the enzyme.
Biochemistry
1995
;
34
:
4202
10
69.
Nackley
AG
,
Shabalina
SA
,
Tchivileva
IE
,
Satterfield
K
,
Korchynskyi
O
,
Makarov
SS
,
Maixner
W
,
Diatchenko
L
:
Human catechol-O-methyltransferase haplotypes modulate protein expression by altering mRNA secondary structure.
Science
2006
;
314
:
1930
3
70.
Slade
GD
,
Sanders
AE
,
Ohrbach
R
,
Bair
E
,
Maixner
W
,
Greenspan
JD
,
Fillingim
RB
,
Smith
S
,
Diatchenko
L
:
COMT diplotype amplifies effect of stress on risk of temporomandibular pain.
J Dent Res
2015
;
94
:
1187
95
71.
Barbosa
FR
,
Matsuda
JB
,
Mazucato
M
,
de Castro França
S
,
Zingaretti
SM
,
da Silva
LM
,
Martinez-Rossi
NM
,
Júnior
MF
,
Marins
M
,
Fachin
AL
:
Influence of catechol-O-methyltransferase (COMT) gene polymorphisms in pain sensibility of Brazilian fibromialgia patients.
Rheumatol Int
2012
;
32
:
427
30
72.
Cohen
H
,
Neumann
L
,
Glazer
Y
,
Ebstein
RP
,
Buskila
D
:
The relationship between a common catechol-O-methyltransferase (COMT) polymorphism val(158) met and fibromyalgia.
Clin Exp Rheumatol
2009
;
27
(
suppl 5
):
S51
6
73.
Gürsoy
S
,
Erdal
E
,
Herken
H
,
Madenci
E
,
Alaşehirli
B
,
Erdal
N
:
Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome.
Rheumatol Int
2003
;
23
:
104
7
74.
Matsuda
JB
,
Barbosa
FR
,
Morel
LJ
,
França
Sde C
,
Zingaretti
SM
,
da Silva
LM
,
Pereira
AM
,
Marins
M
,
Fachin
AL
:
Serotonin receptor (5-HT 2A) and catechol-O-methyltransferase (COMT) gene polymorphisms: Triggers of fibromyalgia?
Rev Bras Reumatol
2010
;
50
:
141
9
75.
Vargas-Alarcón
G
,
Fragoso
JM
,
Cruz-Robles
D
,
Vargas
A
,
Vargas
A
,
Lao-Villadóniga
JI
,
García-Fructuoso
F
,
Ramos-Kuri
M
,
Hernández
F
,
Springall
R
,
Bojalil
R
,
Vallejo
M
,
Martínez-Lavín
M
:
Catechol-O-methyltransferase gene haplotypes in Mexican and Spanish patients with fibromyalgia.
Arthritis Res Ther
2007
;
9
:
R110
76.
Diatchenko
L
,
Slade
GD
,
Nackley
AG
,
Bhalang
K
,
Sigurdsson
A
,
Belfer
I
,
Goldman
D
,
Xu
K
,
Shabalina
SA
,
Shagin
D
,
Max
MB
,
Makarov
SS
,
Maixner
W
:
Genetic basis for individual variations in pain perception and the development of a chronic pain condition.
Hum Mol Genet
2005
;
14
:
135
43
77.
Zubieta
JK
,
Heitzeg
MM
,
Smith
YR
,
Bueller
JA
,
Xu
K
,
Xu
Y
,
Koeppe
RA
,
Stohler
CS
,
Goldman
D
:
COMT val158met genotype affects mu-opioid neurotransmitter responses to a pain stressor.
Science
2003
;
299
:
1240
3
78.
McLean
SA
,
Diatchenko
L
,
Lee
YM
,
Swor
RA
,
Domeier
RM
,
Jones
JS
,
Jones
CW
,
Reed
C
,
Harris
RE
,
Maixner
W
,
Clauw
DJ
,
Liberzon
I
:
Catechol O-methyltransferase haplotype predicts immediate musculoskeletal neck pain and psychological symptoms after motor vehicle collision.
J Pain
2011
;
12
:
101
7
79.
Meloto
CB
,
Bortsov
AV
,
Bair
E
,
Helgeson
E
,
Ostrom
C
,
Smith
SB
,
Dubner
R
,
Slade
GD
,
Fillingim
RB
,
Greenspan
JD
,
Ohrbach
R
,
Maixner
W
,
McLean
SA
,
Diatchenko
L
:
Modification of COMT-dependent pain sensitivity by psychological stress and sex.
Pain
2016
;
157
:
858
67
80.
Hartung
JE
,
Ciszek
BP
,
Nackley
AG
:
β2- and β3-adrenergic receptors drive COMT-dependent pain by increasing production of nitric oxide and cytokines.
Pain
2014
;
155
:
1346
55
81.
Kline
RHt
,
Exposto
FG
,
O’Buckley
SC
,
Westlund
KN
,
Nackley
AG
:
Catechol-O-methyltransferase inhibition alters pain and anxiety-related volitional behaviors through activation of β-adrenergic receptors in the rat.
Neuroscience
2015
;
290
:
561
9
82.
Nackley
AG
,
Tan
KS
,
Fecho
K
,
Flood
P
,
Diatchenko
L
,
Maixner
W
:
Catechol-O-methyltransferase inhibition increases pain sensitivity through activation of both β2- and β3-adrenergic receptors.
Pain
2007
;
128
:
199
208
83.
Ciszek
BP
,
O’Buckley
SC
,
Nackley
AG
:
Persistent catechol-O-methyltransferase-dependent pain is initiated by peripheral β-adrenergic receptors.
Anesthesiology
2016
;
124
:
1122
35
84.
Hartung
J
,
Ciszek
B
,
O’Buckley
S
,
Nackley
A
:
(374) Distinct mechanisms underlie the development and maintenance of COMT-dependent pain.
J Pain
2016
;
17
:
S68
85.
Bote
ME
,
García
JJ
,
Hinchado
MD
,
Ortega
E
:
Inflammatory/stress feedback dysregulation in women with fibromyalgia.
Neuroimmunomodulation
2012
;
19
:
343
51
86.
Larson
AA
,
Giovengo
SL
,
Russell
IJ
,
Michalek
JE
:
Changes in the concentrations of amino acids in the cerebrospinal fluid that correlate with pain in patients with fibromyalgia: Implications for nitric oxide pathways.
Pain
2000
;
87
:
201
11
87.
Shafer
DM
,
Assael
L
,
White
LB
,
Rossomando
EF
:
Tumor necrosis factor-α as a biochemical marker of pain and outcome in temporomandibular joints with internal derangements.
J Oral Maxillofac Surg
1994
;
52
:
786
92
88.
Kubota
E
,
Kubota
T
,
Matsumoto
J
,
Shibata
T
,
Murakami
KI
:
Synovial fluid cytokines and proteinases as markers of temporomandibular joint disease.
J Oral Maxillofac Surg
1998
;
56
:
192
8
89.
Fan
W
,
Huang
F
,
Wu
Z
,
Zhu
X
,
Li
D
,
He
H
:
The role of nitric oxide in orofacial pain.
Nitric Oxide
2012
;
26
:
32
7
90.
Ferrari
LF
,
Araldi
D
,
Levine
JD
:
Distinct terminal and cell body mechanisms in the nociceptor mediate hyperalgesic priming.
J Neurosci
2015
;
35
:
6107
16
91.
Wang
H
,
Heijnen
CJ
,
Eijkelkamp
N
,
Garza Carbajal
A
,
Schedlowski
M
,
Kelley
KW
,
Dantzer
R
,
Kavelaars
A
:
GRK2 in sensory neurons regulates epinephrine-induced signalling and duration of mechanical hyperalgesia.
Pain
2011
;
152
:
1649
58
92.
Ferrari
LF
,
Bogen
O
,
Alessandri-Haber
N
,
Levine
E
,
Gear
RW
,
Levine
JD
:
Transient decrease in nociceptor GRK2 expression produces long-term enhancement in inflammatory pain.
Neuroscience
2012
;
222
:
392
403
93.
Eijkelkamp
N
,
Wang
H
,
Garza-Carbajal
A
,
Willemen
HL
,
Zwartkruis
FJ
,
Wood
JN
,
Dantzer
R
,
Kelley
KW
,
Heijnen
CJ
,
Kavelaars
A
:
Low nociceptor GRK2 prolongs prostaglandin E2 hyperalgesia via biased cAMP signaling to Epac/Rap1, protein kinase Cε, and MEK/ERK.
J Neurosci
2010
;
30
:
12806
15
94.
McLean
SA
:
The potential contribution of stress systems to the transition to chronic whiplash-associated disorders.
Spine (Phila Pa 1976)
2011
;
36
(
suppl 25
):
S226
32
95.
Khasar
SG
,
Lin
YH
,
Martin
A
,
Dadgar
J
,
McMahon
T
,
Wang
D
,
Hundle
B
,
Aley
KO
,
Isenberg
W
,
McCarter
G
,
Green
PG
,
Hodge
CW
,
Levine
JD
,
Messing
RO
:
A novel nociceptor signaling pathway revealed in protein kinase Cε mutant mice.
Neuron
1999
;
24
:
253
60
96.
Khasar
SG
,
McCarter
G
,
Levine
JD
:
Epinephrine produces a beta-adrenergic receptor-mediated mechanical hyperalgesia and in vitro sensitization of rat nociceptors.
J Neurophysiol
1999
;
81
:
1104
12
97.
Akimoto
Y
,
Horinouchi
T
,
Shibano
M
,
Matsushita
M
,
Yamashita
Y
,
Okamoto
T
,
Yamaki
F
,
Tanaka
Y
,
Koike
K
:
Nitric oxide (NO) primarily accounts for endothelium-dependent component of β-adrenoceptor-activated smooth muscle relaxation of mouse aorta in response to isoprenaline.
J Smooth Muscle Res
2002
;
38
:
87
99
98.
Canová
NK
,
Lincová
D
,
Kmonícková
E
,
Kameníková
L
,
Farghali
H
:
Nitric oxide production from rat adipocytes is modulated by β3-adrenergic receptor agonists and is involved in a cyclic AMP-dependent lipolysis in adipocytes.
Nitric Oxide
2006
;
14
:
200
11
99.
Figueroa
XF
,
Poblete
I
,
Fernández
R
,
Pedemonte
C
,
Cortés
V
,
Huidobro-Toro
JP
:
NO production and eNOS phosphorylation induced by epinephrine through the activation of β-adrenoceptors.
Am J Physiol Heart Circ Physiol
2009
;
297
:
H134
43
100.
Frost
RA
,
Nystrom
GJ
,
Lang
CH
:
Epinephrine stimulates IL-6 expression in skeletal muscle and C2C12 myoblasts: Role of c-Jun NH2-terminal kinase and histone deacetylase activity.
Am J Physiol Endocrinol Metab
2004
;
286
:
E809
17
101.
Furlán
C
,
Sterin-Borda
L
,
Borda
E
:
Activation of β3 adrenergic receptor decreases DNA synthesis in human skin fibroblasts via cyclic GMP/nitric oxide pathway.
Cell Physiol Biochem
2005
;
16
:
175
82
102.
Hodges
GJ
,
Kosiba
WA
,
Zhao
K
,
Johnson
JM
:
The involvement of norepinephrine, neuropeptide Y, and nitric oxide in the cutaneous vasodilator response to local heating in humans.
J Appl Physiol (1985)
2008
;
105
:
233
40
103.
Maimone
D
,
Cioni
C
,
Rosa
S
,
Macchia
G
,
Aloisi
F
,
Annunziata
P
:
Norepinephrine and vasoactive intestinal peptide induce IL-6 secretion by astrocytes: Synergism with IL-1 β and TNF α.
J Neuroimmunol
1993
;
47
:
73
81
104.
Tchivileva
IE
,
Tan
KS
,
Gambarian
M
,
Nackley
AG
,
Medvedev
AV
,
Romanov
S
,
Flood
PM
,
Maixner
W
,
Makarov
SS
,
Diatchenko
L
:
Signaling pathways mediating β3-adrenergic receptor-induced production of interleukin-6 in adipocytes.
Mol Immunol
2009
;
46
:
2256
66
105.
Tan
KS
,
Nackley
AG
,
Satterfield
K
,
Maixner
W
,
Diatchenko
L
,
Flood
PM
:
β2 adrenergic receptor activation stimulates pro-inflammatory cytokine production in macrophages via PKA- and NF-κB-independent mechanisms.
Cell Signal
2007
;
19
:
251
60
106.
Hains
LE
,
Loram
LC
,
Weiseler
JL
,
Frank
MG
,
Bloss
EB
,
Sholar
P
,
Taylor
FR
,
Harrison
JA
,
Martin
TJ
,
Eisenach
JC
,
Maier
SF
,
Watkins
LR
:
Pain intensity and duration can be enhanced by prior challenge: Initial evidence suggestive of a role of microglial priming.
J Pain
2010
;
11
:
1004
14
107.
White
FA
,
Bhangoo
SK
,
Miller
RJ
:
Chemokines: Integrators of pain and inflammation.
Nat Rev Drug Discov
2005
;
4
:
834
44
108.
Ji
RR
,
Berta
T
,
Nedergaard
M
:
Glia and pain: Is chronic pain a gliopathy?
Pain
2013
;
154
(
suppl 1
):
S10
28
109.
Tsuda
M
,
Inoue
K
,
Salter
MW
:
Neuropathic pain and spinal microglia: A big problem from molecules in “small” glia.
Trends Neurosci
2005
;
28
:
101
7
110.
Ren
K
,
Dubner
R
:
Interactions between the immune and nervous systems in pain.
Nat Med
2010
;
16
:
1267
76
111.
Milligan
ED
,
Watkins
LR
:
Pathological and protective roles of glia in chronic pain.
Nat Rev Neurosci
2009
;
10
:
23
36
112.
McMahon
SB
,
Malcangio
M
:
Current challenges in glia-pain biology.
Neuron
2009
;
64
:
46
54
113.
Clark
AK
,
Yip
PK
,
Grist
J
,
Gentry
C
,
Staniland
AA
,
Marchand
F
,
Dehvari
M
,
Wotherspoon
G
,
Winter
J
,
Ullah
J
,
Bevan
S
,
Malcangio
M
:
Inhibition of spinal microglial cathepsin S for the reversal of neuropathic pain.
Proc Natl Acad Sci USA
2007
;
104
:
10655
60
114.
Garrison
CJ
,
Dougherty
PM
,
Kajander
KC
,
Carlton
SM
:
Staining of glial fibrillary acidic protein (GFAP) in lumbar spinal cord increases following a sciatic nerve constriction injury.
Brain Res
1991
;
565
:
1
7
115.
Grace
PM
,
Strand
KA
,
Galer
EL
,
Urban
DJ
,
Wang
X
,
Baratta
MV
,
Fabisiak
TJ
,
Anderson
ND
,
Cheng
K
,
Greene
LI
,
Berkelhammer
D
,
Zhang
Y
,
Ellis
AL
,
Yin
HH
,
Campeau
S
,
Rice
KC
,
Roth
BL
,
Maier
SF
,
Watkins
LR
:
Morphine paradoxically prolongs neuropathic pain in rats by amplifying spinal NLRP3 inflammasome activation.
Proc Natl Acad Sci USA
2016
;
113
:
E3441
50
116.
Gao
YJ
,
Ji
RR
:
Targeting astrocyte signaling for chronic pain.
Neurotherapeutics
2010
;
7
:
482
93
117.
Fu
KY
,
Light
AR
,
Matsushima
GK
,
Maixner
W
:
Microglial reactions after subcutaneous formalin injection into the rat hind paw.
Brain Res
1999
;
825
:
59
67
118.
Loggia
ML
,
Chonde
DB
,
Akeju
O
,
Arabasz
G
,
Catana
C
,
Edwards
RR
,
Hill
E
,
Hsu
S
,
Izquierdo-Garcia
D
,
Ji
RR
,
Riley
M
,
Wasan
AD
,
Zürcher
NR
,
Albrecht
DS
,
Vangel
MG
,
Rosen
BR
,
Napadow
V
,
Hooker
JM
:
Evidence for brain glial activation in chronic pain patients.
Brain
2015
;
138
:
604
15
119.
Zhu
X
,
Conklin
D
,
Eisenach
JC
:
Cyclooxygenase-1 in the spinal cord plays an important role in postoperative pain.
Pain
2003
;
104
:
15
23
120.
Zhu
X
,
Eisenach
JC
:
Cyclooxygenase-1 in the spinal cord is altered after peripheral nerve injury.
Anesthesiology
2003
;
99
:
1175
9
121.
Guan
Z
,
Kuhn
JA
,
Wang
X
,
Colquitt
B
,
Solorzano
C
,
Vaman
S
,
Guan
AK
,
Evans-Reinsch
Z
,
Braz
J
,
Devor
M
,
Abboud-Werner
SL
,
Lanier
LL
,
Lomvardas
S
,
Basbaum
AI
:
Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain.
Nat Neurosci
2016
;
19
:
94
101
122.
Jiang
BC
,
Cao
DL
,
Zhang
X
,
Zhang
ZJ
,
He
LN
,
Li
CH
,
Zhang
WW
,
Wu
XB
,
Berta
T
,
Ji
RR
,
Gao
YJ
:
CXCL13 drives spinal astrocyte activation and neuropathic pain via CXCR5.
J Clin Invest
2016
;
126
:
745
61
123.
Wu
Y
,
Willcockson
HH
,
Maixner
W
,
Light
AR
:
Suramin inhibits spinal cord microglia activation and long-term hyperalgesia induced by formalin injection.
J Pain
2004
;
5
:
48
55
124.
Tsuda
M
,
Shigemoto-Mogami
Y
,
Koizumi
S
,
Mizokoshi
A
,
Kohsaka
S
,
Salter
MW
,
Inoue
K
:
P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury.
Nature
2003
;
424
:
778
83
125.
Tsuda
M
,
Tozaki-Saitoh
H
,
Inoue
K
:
Pain and purinergic signaling.
Brain Res Rev
2010
;
63
:
222
32
126.
Trang
T
,
Beggs
S
,
Salter
MW
:
ATP receptors gate microglia signaling in neuropathic pain.
Exp Neurol
2012
;
234
:
354
61
127.
Kang
J
,
Kang
N
,
Lovatt
D
,
Torres
A
,
Zhao
Z
,
Lin
J
,
Nedergaard
M
:
Connexin 43 hemichannels are permeable to ATP.
J Neurosci
2008
;
28
:
4702
11
128.
Koyanagi
S
,
Kusunose
N
,
Taniguchi
M
,
Akamine
T
,
Kanado
Y
,
Ozono
Y
,
Masuda
T
,
Kohro
Y
,
Matsunaga
N
,
Tsuda
M
,
Salter
MW
,
Inoue
K
,
Ohdo
S
:
Glucocorticoid regulation of ATP release from spinal astrocytes underlies diurnal exacerbation of neuropathic mechanical allodynia.
Nat Commun
2016
;
7
:
13102
129.
Masuda
T
,
Ozono
Y
,
Mikuriya
S
,
Kohro
Y
,
Tozaki-Saitoh
H
,
Iwatsuki
K
,
Uneyama
H
,
Ichikawa
R
,
Salter
MW
,
Tsuda
M
,
Inoue
K
:
Dorsal horn neurons release extracellular ATP in a VNUT-dependent manner that underlies neuropathic pain.
Nat Commun
2016
;
7
:
12529
130.
Burma
NE
,
Bonin
RP
,
Leduc-Pessah
H
,
Baimel
C
,
Cairncross
ZF
,
Mousseau
M
,
Shankara
JV
,
Stemkowski
PL
,
Baimoukhametova
D
,
Bains
JS
,
Antle
MC
,
Zamponi
GW
,
Cahill
CM
,
Borgland
SL
,
De Koninck
Y
,
Trang
T
:
Blocking microglial pannexin-1 channels alleviates morphine withdrawal in rodents.
Nat Med
2017
;
23
:
355
60
131.
Milligan
ED
,
Zapata
V
,
Chacur
M
,
Schoeniger
D
,
Biedenkapp
J
,
O’Connor
KA
,
Verge
GM
,
Chapman
G
,
Green
P
,
Foster
AC
,
Naeve
GS
,
Maier
SF
,
Watkins
LR
:
Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats.
Eur J Neurosci
2004
;
20
:
2294
302
132.
Zhuang
ZY
,
Kawasaki
Y
,
Tan
PH
,
Wen
YR
,
Huang
J
,
Ji
RR
:
Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine.
Brain Behav Immun
2007
;
21
:
642
51
133.
Yang
Y
,
Li
H
,
Li
TT
,
Luo
H
,
Gu
XY
,
N
,
Ji
RR
,
Zhang
YQ
:
Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18.
J Neurosci
2015
;
35
:
7950
63
134.
Zhang
MD
,
Barde
S
,
Yang
T
,
Lei
B
,
Eriksson
LI
,
Mathew
JP
,
Andreska
T
,
Akassoglou
K
,
Harkany
T
,
Hökfelt
TG
,
Terrando
N
:
Orthopedic surgery modulates neuropeptides and BDNF expression at the spinal and hippocampal levels.
Proc Natl Acad Sci USA
2016
;
113
:
E6686
95
135.
Li
WW
,
Guo
TZ
,
Shi
X
,
Sun
Y
,
Wei
T
,
Clark
DJ
,
Kingery
WS
:
Substance P spinal signaling induces glial activation and nociceptive sensitization after fracture.
Neuroscience
2015
;
310
:
73
90
136.
Sung
B
,
Lim
G
,
Mao
J
:
Altered expression and uptake activity of spinal glutamate transporters after nerve injury contribute to the pathogenesis of neuropathic pain in rats.
J Neurosci
2003
;
23
:
2899
910
137.
Cotrina
ML
,
Lin
JH
,
López-García
JC
,
Naus
CC
,
Nedergaard
M
:
ATP-mediated glia signaling.
J Neurosci
2000
;
20
:
2835
44
138.
Chen
MJ
,
Kress
B
,
Han
X
,
Moll
K
,
Peng
W
,
Ji
RR
,
Nedergaard
M
:
Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury.
Glia
2012
;
60
:
1660
70
139.
Chen
G
,
Park
CK
,
Xie
RG
,
Berta
T
,
Nedergaard
M
,
Ji
RR
:
Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice.
Brain
2014
;
137
:
2193
209
140.
Ren
K
,
Torres
R
:
Role of interleukin-1β during pain and inflammation.
Brain Res Rev
2009
;
60
:
57
64
141.
Gao
YJ
,
Ji
RR
:
Chemokines, neuronal-glial interactions, and central processing of neuropathic pain.
Pharmacol Ther
2010
;
126
:
56
68
142.
Kim
DS
,
Li
KW
,
Boroujerdi
A
,
Peter Yu
Y
,
Zhou
CY
,
Deng
P
,
Park
J
,
Zhang
X
,
Lee
J
,
Corpe
M
,
Sharp
K
,
Steward
O
,
Eroglu
C
,
Barres
B
,
Zaucke
F
,
Xu
ZC
,
Luo
ZD
:
Thrombospondin-4 contributes to spinal sensitization and neuropathic pain states.
J Neurosci
2012
;
32
:
8977
87
143.
Ji
RR
,
Gereau
RW
IV
,
Malcangio
M
,
Strichartz
GR
:
MAP kinase and pain.
Brain Res Rev
2009
;
60
:
135
48
144.
Ji
RR
,
Suter
MR
:
p38 MAPK, microglial signaling, and neuropathic pain.
Mol Pain
2007
;
3
:
33
145.
Trang
T
,
Beggs
S
,
Wan
X
,
Salter
MW
:
P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation.
J Neurosci
2009
;
29
:
3518
28
146.
Kobayashi
K
,
Yamanaka
H
,
Fukuoka
T
,
Dai
Y
,
Obata
K
,
Noguchi
K
:
P2Y12 receptor upregulation in activated microglia is a gateway of p38 signaling and neuropathic pain.
J Neurosci
2008
;
28
:
2892
902
147.
Clark
AK
,
Staniland
AA
,
Marchand
F
,
Kaan
TK
,
McMahon
SB
,
Malcangio
M
:
P2X7-dependent release of interleukin-1β and nociception in the spinal cord following lipopolysaccharide.
J Neurosci
2010
;
30
:
573
82
148.
Zhuang
ZY
,
Wen
YR
,
Zhang
DR
,
Borsello
T
,
Bonny
C
,
Strichartz
GR
,
Decosterd
I
,
Ji
RR
:
A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: Respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance.
J Neurosci
2006
;
26
:
3551
60
149.
Gao
YJ
,
Xu
ZZ
,
Liu
YC
,
Wen
YR
,
Decosterd
I
,
Ji
RR
:
The c-Jun N-terminal kinase 1 (JNK1) in spinal astrocytes is required for the maintenance of bilateral mechanical allodynia under a persistent inflammatory pain condition.
Pain
2010
;
148
:
309
19
150.
Gao
YJ
,
Zhang
L
,
Samad
OA
,
Suter
MR
,
Yasuhiko
K
,
Xu
ZZ
,
Park
JY
,
Lind
AL
,
Ma
Q
,
Ji
RR
:
JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain.
J Neurosci
2009
;
29
:
4096
108
151.
Kim
SK
,
Hayashi
H
,
Ishikawa
T
,
Shibata
K
,
Shigetomi
E
,
Shinozaki
Y
,
Inada
H
,
Roh
SE
,
Kim
SJ
,
Lee
G
,
Bae
H
,
Moorhouse
AJ
,
Mikoshiba
K
,
Fukazawa
Y
,
Koizumi
S
,
Nabekura
J
:
Cortical astrocytes rewire somatosensory cortical circuits for peripheral neuropathic pain.
J Clin Invest
2016
;
126
:
1983
97
152.
Tanga
FY
,
Nutile-McMenemy
N
,
DeLeo
JA
:
The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy.
Proc Natl Acad Sci USA
2005
;
102
:
5856
61
153.
Liu
T
,
Han
Q
,
Chen
G
,
Huang
Y
,
Zhao
LX
,
Berta
T
,
Gao
YJ
,
Ji
RR
:
Toll-like receptor 4 contributes to chronic itch, alloknesis, and spinal astrocyte activation in male mice.
Pain
2016
;
157
:
806
17
154.
Tramullas
M
,
Finger
BC
,
Moloney
RD
,
Golubeva
AV
,
Moloney
G
,
Dinan
TG
,
Cryan
JF
:
Toll-like receptor 4 regulates chronic stress-induced visceral pain in mice.
Biol Psychiatry
2014
;
76
:
340
8
155.
Taylor
AM
,
Castonguay
A
,
Taylor
AJ
,
Murphy
NP
,
Ghogha
A
,
Cook
C
,
Xue
L
,
Olmstead
MC
,
De Koninck
Y
,
Evans
CJ
,
Cahill
CM
:
Microglia disrupt mesolimbic reward circuitry in chronic pain.
J Neurosci
2015
;
35
:
8442
50
156.
Zhang
F
,
Vadakkan
KI
,
Kim
SS
,
Wu
LJ
,
Shang
Y
,
Zhuo
M
:
Selective activation of microglia in spinal cord but not higher cortical regions following nerve injury in adult mouse.
Mol Pain
2008
;
4
:
15
157.
Tsuda
M
,
Koga
K
,
Chen
T
,
Zhuo
M
:
Neuronal and microglial mechanisms for neuropathic pain in the spinal dorsal horn and anterior cingulate cortex.
J Neurochem
2017
;
141
:
486
98
158.
Horst
OV
,
Cunha-Cruz
J
,
Zhou
L
,
Manning
W
,
Mancl
L
,
DeRouen
TA
:
Prevalence of pain in the orofacial regions in patients visiting general dentists in the Northwest Practice-based REsearch Collaborative in Evidence-based DENTistry research network.
J Am Dent Assoc
2015
;
146
:
721
8.e3
159.
Slade
GD
,
Fillingim
RB
,
Sanders
AE
,
Bair
E
,
Greenspan
JD
,
Ohrbach
R
,
Dubner
R
,
Diatchenko
L
,
Smith
SB
,
Knott
C
,
Maixner
W
:
Summary of findings from the OPPERA prospective cohort study of incidence of first-onset temporomandibular disorder: Implications and future directions.
J Pain
2013
;
14
(
suppl 12
):
T116
24
160.
Maixner
W
,
Humphrey
C
:
Gender differences in pain and cardiovascular responses to forearm ischemia.
Clin J Pain
1993
;
9
:
16
25
161.
Mogil
JS
:
Sex differences in pain and pain inhibition: Multiple explanations of a controversial phenomenon.
Nat Rev Neurosci
2012
;
13
:
859
66
162.
Sorge
RE
,
LaCroix-Fralish
ML
,
Tuttle
AH
,
Sotocinal
SG
,
Austin
JS
,
Ritchie
J
,
Chanda
ML
,
Graham
AC
,
Topham
L
,
Beggs
S
,
Salter
MW
,
Mogil
JS
:
Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice.
J Neurosci
2011
;
31
:
15450
4
163.
Sorge
RE
,
Mapplebeck
JC
,
Rosen
S
,
Beggs
S
,
Taves
S
,
Alexander
JK
,
Martin
LJ
,
Austin
JS
,
Sotocinal
SG
,
Chen
D
,
Yang
M
,
Shi
XQ
,
Huang
H
,
Pillon
NJ
,
Bilan
PJ
,
Tu
Y
,
Klip
A
,
Ji
RR
,
Zhang
J
,
Salter
MW
,
Mogil
JS
:
Different immune cells mediate mechanical pain hypersensitivity in male and female mice.
Nat Neurosci
2015
;
18
:
1081
3
164.
Taves
S
,
Berta
T
,
Liu
DL
,
Gan
S
,
Chen
G
,
Kim
YH
,
Van de Ven
T
,
Laufer
S
,
Ji
RR
:
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.
Brain Behav Immun
2016
;
55
:
70
81
165.
Berta
T
,
Qadri
YJ
,
Chen
G
,
Ji
RR
:
Microglial signaling in chronic pain with a special focus on caspase 6, p38 MAP kinase, and sex dependence.
J Dent Res
2016
;
95
:
1124
31
166.
Berta
T
,
Park
CK
,
Xu
ZZ
,
Xie
RG
,
Liu
T
,
N
,
Liu
YC
,
Ji
RR
:
Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion.
J Clin Invest
2014
;
124
:
1173
86
167.
Clauw
DJ
:
Diagnosing and treating chronic musculoskeletal pain based on the underlying mechanism(s).
Best Pract Res Clin Rheumatol
2015
;
29
:
6
19
168.
Yunus
MB
:
Editorial review: An update on central sensitivity syndromes and the issues of nosology and psychobiology.
Curr Rheumatol Rev
2015
;
11
:
70
85
169.
Loeser
JD
,
Treede
RD
:
The Kyoto protocol of IASP Basic Pain Terminology.
Pain
2008
;
137
:
473
7
170.
Ren
K
,
Hylden
JL
,
Williams
GM
,
Ruda
MA
,
Dubner
R
:
The effects of a non-competitive NMDA receptor antagonist, MK-801, on behavioral hyperalgesia and dorsal horn neuronal activity in rats with unilateral inflammation.
Pain
1992
;
50
:
331
44
171.
Dubner
R
,
Ruda
MA
:
Activity-dependent neuronal plasticity following tissue injury and inflammation.
Trends Neurosci
1992
;
15
:
96
103
172.
Liu
XJ
,
Gingrich
JR
,
Vargas-Caballero
M
,
Dong
YN
,
Sengar
A
,
Beggs
S
,
Wang
SH
,
Ding
HK
,
Frankland
PW
,
Salter
MW
:
Treatment of inflammatory and neuropathic pain by uncoupling Src from the NMDA receptor complex.
Nat Med
2008
;
14
:
1325
32
173.
Chen
G
,
Xie
RG
,
Gao
YJ
,
Xu
ZZ
,
Zhao
LX
,
Bang
S
,
Berta
T
,
Park
CK
,
Lay
M
,
Chen
W
,
Ji
RR
:
β-Arrestin-2 regulates NMDA receptor function in spinal lamina II neurons and duration of persistent pain.
Nat Commun
2016
;
7
:
12531
174.
Bohn
LM
,
Lefkowitz
RJ
,
Gainetdinov
RR
,
Peppel
K
,
Caron
MG
,
Lin
FT
:
Enhanced morphine analgesia in mice lacking β-arrestin 2.
Science
1999
;
286
:
2495
8
175.
Hartmann
B
,
Ahmadi
S
,
Heppenstall
PA
,
Lewin
GR
,
Schott
C
,
Borchardt
T
,
Seeburg
PH
,
Zeilhofer
HU
,
Sprengel
R
,
Kuner
R
:
The AMPA receptor subunits GluR-A and GluR-B reciprocally modulate spinal synaptic plasticity and inflammatory pain.
Neuron
2004
;
44
:
637
50
176.
Tao
YX
:
AMPA receptor trafficking in inflammation-induced dorsal horn central sensitization.
Neurosci Bull
2012
;
28
:
111
20
177.
Tappe
A
,
Klugmann
M
,
Luo
C
,
Hirlinger
D
,
Agarwal
N
,
Benrath
J
,
Ehrengruber
MU
,
During
MJ
,
Kuner
R
:
Synaptic scaffolding protein Homer1a protects against chronic inflammatory pain.
Nat Med
2006
;
12
:
677
81
178.
Zhang
WB
,
Ross
PJ
,
Tu
Y
,
Wang
Y
,
Beggs
S
,
Sengar
AS
,
Ellis
J
,
Salter
MW
:
Fyn Kinase regulates GluN2B subunit-dominant NMDA receptors in human induced pluripotent stem cell-derived neurons.
Sci Rep
2016
;
6
:
23837
179.
Ji
RR
,
Baba
H
,
Brenner
GJ
,
Woolf
CJ
:
Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity.
Nat Neurosci
1999
;
2
:
1114
9
180.
Karim
F
,
Wang
CC
,
Gereau
RW
IV
:
Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal-regulated kinase signaling required for inflammatory pain in mice.
J Neurosci
2001
;
21
:
3771
9
181.
Gao
YJ
,
Ji
RR
:
c-Fos and pERK, which is a better marker for neuronal activation and central sensitization after noxious stimulation and tissue injury?
Open Pain J
2009
;
2
:
11
7
182.
Kawasaki
Y
,
Kohno
T
,
Zhuang
ZY
,
Brenner
GJ
,
Wang
H
,
Van Der Meer
C
,
Befort
K
,
Woolf
CJ
,
Ji
RR
:
Ionotropic and metabotropic receptors, protein kinase A, protein kinase C, and Src contribute to C-fiber-induced ERK activation and cAMP response element-binding protein phosphorylation in dorsal horn neurons, leading to central sensitization.
J Neurosci
2004
;
24
:
8310
21
183.
Hu
HJ
,
Gereau
RW
IV
:
ERK integrates PKA and PKC signaling in superficial dorsal horn neurons: II. Modulation of neuronal excitability.
J Neurophysiol
2003
;
90
:
1680
8
184.
Wei
F
,
Vadakkan
KI
,
Toyoda
H
,
Wu
LJ
,
Zhao
MG
,
Xu
H
,
Shum
FW
,
Jia
YH
,
Zhuo
M
:
Calcium calmodulin-stimulated adenylyl cyclases contribute to activation of extracellular signal-regulated kinase in spinal dorsal horn neurons in adult rats and mice.
J Neurosci
2006
;
26
:
851
61
185.
Hu
HJ
,
Glauner
KS
,
Gereau
RW
IV
:
ERK integrates PKA and PKC signaling in superficial dorsal horn neurons: I. Modulation of A-type K+ currents.
J Neurophysiol
2003
;
90
:
1671
9
186.
Hu
HJ
,
Carrasquillo
Y
,
Karim
F
,
Jung
WE
,
Nerbonne
JM
,
Schwarz
TL
,
Gereau
RW
IV
:
The kv4.2 potassium channel subunit is required for pain plasticity.
Neuron
2006
;
50
:
89
100
187.
Xu
ZZ
,
Zhang
L
,
Liu
T
,
Park
JY
,
Berta
T
,
Yang
R
,
Serhan
CN
,
Ji
RR
:
Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions.
Nat Med
2010
;
16
:
592
7
188.
Ji
RR
,
Befort
K
,
Brenner
GJ
,
Woolf
CJ
:
ERK MAP kinase activation in superficial spinal cord neurons induces prodynorphin and NK-1 upregulation and contributes to persistent inflammatory pain hypersensitivity.
J Neurosci
2002
;
22
:
478
85
189.
Baba
H
,
Ji
RR
,
Kohno
T
,
Moore
KA
,
Ataka
T
,
Wakai
A
,
Okamoto
M
,
Woolf
CJ
:
Removal of GABAergic inhibition facilitates polysynaptic A fiber-mediated excitatory transmission to the superficial spinal dorsal horn.
Mol Cell Neurosci
2003
;
24
:
818
30
190.
Melzack
R
,
Wall
PD
:
Pain mechanisms: A new theory.
Science
1965
;
150
:
971
9
191.
Hains
BC
,
Waxman
SG
:
Activated microglia contribute to the maintenance of chronic pain after spinal cord injury.
J Neurosci
2006
;
26
:
4308
17
192.
Yang
Q
,
Wu
Z
,
Hadden
JK
,
Odem
MA
,
Zuo
Y
,
Crook
RJ
,
Frost
JA
,
Walters
ET
:
Persistent pain after spinal cord injury is maintained by primary afferent activity.
J Neurosci
2014
;
34
:
10765
9
193.
Samad
TA
,
Moore
KA
,
Sapirstein
A
,
Billet
S
,
Allchorne
A
,
Poole
S
,
Bonventre
JV
,
Woolf
CJ
:
Interleukin-1β-mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity.
Nature
2001
;
410
:
471
5
194.
Kawasaki
Y
,
Zhang
L
,
Cheng
JK
,
Ji
RR
:
Cytokine mechanisms of central sensitization: Distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord.
J Neurosci
2008
;
28
:
5189
94
195.
Zhou
LJ
,
Liu
XG
:
Glial activation, a common mechanism underlying spinal synaptic plasticity?
Neurosci Bull
2017
;
33
:
121
3
196.
Wang
L
,
Bauer
M
,
Curry
R
,
Larsson
A
,
Sessler
DI
,
Eisenach
JC
:
Intrathecal ketorolac does not improve acute or chronic pain after hip arthroplasty: A randomized controlled trial.
J Anesth
2014
;
28
:
790
3
197.
Ahmadi
S
,
Lippross
S
,
Neuhuber
WL
,
Zeilhofer
HU
:
PGE2 selectively blocks inhibitory glycinergic neurotransmission onto rat superficial dorsal horn neurons.
Nat Neurosci
2002
;
5
:
34
40
198.
DeLeo
JA
,
Tanga
FY
,
Tawfik
VL
:
Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia.
Neuroscientist
2004
;
10
:
40
52
199.
Milligan
ED
,
Twining
C
,
Chacur
M
,
Biedenkapp
J
,
O’Connor
K
,
Poole
S
,
Tracey
K
,
Martin
D
,
Maier
SF
,
Watkins
LR
:
Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain in rats.
J Neurosci
2003
;
23
:
1026
40
200.
Schäfers
M
,
Geis
C
,
Svensson
CI
,
Luo
ZD
,
Sommer
C
:
Selective increase of tumour necrosis factor-α in injured and spared myelinated primary afferents after chronic constrictive injury of rat sciatic nerve.
Eur J Neurosci
2003
;
17
:
791
804
201.
Xu
JT
,
Xin
WJ
,
Zang
Y
,
Wu
CY
,
Liu
XG
:
The role of tumor necrosis factor-α in the neuropathic pain induced by Lumbar 5 ventral root transection in rat.
Pain
2006
;
123
:
306
21
202.
Park
CK
,
N
,
Xu
ZZ
,
Liu
T
,
Serhan
CN
,
Ji
RR
:
Resolving TRPV1- and TNF-α-mediated spinal cord synaptic plasticity and inflammatory pain with neuroprotectin D1.
J Neurosci
2011
;
31
:
15072
85
203.
Zhang
H
,
Nei
H
,
Dougherty
PM
:
A p38 mitogen-activated protein kinase-dependent mechanism of disinhibition in spinal synaptic transmission induced by tumor necrosis factor-α.
J Neurosci
2010
;
30
:
12844
55
204.
Zhang
L
,
Berta
T
,
Xu
ZZ
,
Liu
T
,
Park
JY
,
Ji
RR
:
TNF-α contributes to spinal cord synaptic plasticity and inflammatory pain: Distinct role of TNF receptor subtypes 1 and 2.
Pain
2011
;
152
:
419
27
205.
Zhang
RX
,
Liu
B
,
Wang
L
,
Ren
K
,
Qiao
JT
,
Berman
BM
,
Lao
L
:
Spinal glial activation in a new rat model of bone cancer pain produced by prostate cancer cell inoculation of the tibia.
Pain
2005
;
118
:
125
36
206.
Miyoshi
K
,
Obata
K
,
Kondo
T
,
Okamura
H
,
Noguchi
K
:
Interleukin-18-mediated microglia/astrocyte interaction in the spinal cord enhances neuropathic pain processing after nerve injury.
J Neurosci
2008
;
28
:
12775
87
207.
Li
WW
,
Guo
TZ
,
Liang
D
,
Shi
X
,
Wei
T
,
Kingery
WS
,
Clark
JD
:
The NALP1 inflammasome controls cytokine production and nociception in a rat fracture model of complex regional pain syndrome.
Pain
2009
;
147
:
277
86
208.
Kawasaki
Y
,
Xu
ZZ
,
Wang
X
,
Park
JY
,
Zhuang
ZY
,
Tan
PH
,
Gao
YJ
,
Roy
K
,
Corfas
G
,
Lo
EH
,
Ji
RR
:
Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain.
Nat Med
2008
;
14
:
331
6
209.
Guo
W
,
Wang
H
,
Watanabe
M
,
Shimizu
K
,
Zou
S
,
LaGraize
SC
,
Wei
F
,
Dubner
R
,
Ren
K
:
Glial-cytokine-neuronal interactions underlying the mechanisms of persistent pain.
J Neurosci
2007
;
27
:
6006
18
210.
Zhang
RX
,
Li
A
,
Liu
B
,
Wang
L
,
Ren
K
,
Zhang
H
,
Berman
BM
,
Lao
L
:
IL-1ra alleviates inflammatory hyperalgesia through preventing phosphorylation of NMDA receptor NR-1 subunit in rats.
Pain
2008
;
135
:
232
9
211.
Yan
X
,
Weng
HR
:
Endogenous interleukin-1β in neuropathic rats enhances glutamate release from the primary afferents in the spinal dorsal horn through coupling with presynaptic NMDA receptors.
J Biol Chem
2013
;
288
:
30544
57
212.
Zhang
ZJ
,
Cao
DL
,
Zhang
X
,
Ji
RR
,
Gao
YJ
:
Chemokine contribution to neuropathic pain: Respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons.
Pain
2013
;
154
:
2185
97
213.
White
FA
,
Sun
J
,
Waters
SM
,
Ma
C
,
Ren
D
,
Ripsch
M
,
Steflik
J
,
Cortright
DN
,
Lamotte
RH
,
Miller
RJ
:
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
7
214.
Xie
RG
,
Gao
YJ
,
Park
CK
,
Lu
N
,
Luo
C
,
Wang
WT
,
Wu
SX
,
Ji
RR
:
Spinal CCL2 promotes central sensitization, long-term potentiation, and inflammatory pain via CCR2: Further insights into molecular, synaptic, and cellular mechanisms.
Neurosci Bull
2018
;
34
:
13
21
215.
Sun
Y
,
Sahbaie
P
,
Liang
DY
,
Li
WW
,
Li
XQ
,
Shi
XY
,
Clark
JD
:
Epigenetic regulation of spinal CXCR2 signaling in incisional hypersensitivity in mice.
Anesthesiology
2013
;
119
:
1198
208
216.
Coull
JA
,
Beggs
S
,
Boudreau
D
,
Boivin
D
,
Tsuda
M
,
Inoue
K
,
Gravel
C
,
Salter
MW
,
De Koninck
Y
:
BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain.
Nature
2005
;
438
:
1017
21
217.
Ji
RR
,
Kawasaki
Y
,
Zhuang
ZY
,
Wen
YR
,
Decosterd
I
:
Possible role of spinal astrocytes in maintaining chronic pain sensitization: Review of current evidence with focus on bFGF/JNK pathway.
Neuron Glia Biol
2006
;
2
:
259
69
218.
Fukuoka
T
,
Kondo
E
,
Dai
Y
,
Hashimoto
N
,
Noguchi
K
:
Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model.
J Neurosci
2001
;
21
:
4891
900
219.
Lever
IJ
,
Bradbury
EJ
,
Cunningham
JR
,
Adelson
DW
,
Jones
MG
,
McMahon
SB
,
Marvizón
JC
,
Malcangio
M
:
Brain-derived neurotrophic factor is released in the dorsal horn by distinctive patterns of afferent fiber stimulation.
J Neurosci
2001
;
21
:
4469
77
220.
Slack
SE
,
Pezet
S
,
McMahon
SB
,
Thompson
SW
,
Malcangio
M
:
Brain-derived neurotrophic factor induces NMDA receptor subunit one phosphorylation via ERK and PKC in the rat spinal cord.
Eur J Neurosci
2004
;
20
:
1769
78
221.
Ferrini
F
,
Trang
T
,
Mattioli
TA
,
Laffray
S
,
Del’guidice
T
,
Lorenzo
LE
,
Castonguay
A
,
Doyon
N
,
Zhang
W
,
Godin
AG
,
Mohr
D
,
Beggs
S
,
Vandal
K
,
Beaulieu
JM
,
Cahill
CM
,
Salter
MW
,
De
KY
:
Morphine hyperalgesia gated through microglia-mediated disruption of neuronal Cl homeostasis.
Nat Neurosci
2013
;
16
:
183
92
222.
Sandkühler
J
:
Learning and memory in pain pathways.
Pain
2000
;
88
:
113
8
223.
Sandkühler
J
:
Models and mechanisms of hyperalgesia and allodynia.
Physiol Rev
2009
;
89
:
707
58
224.
Ruscheweyh
R
,
Wilder-Smith
O
,
Drdla
R
,
Liu
XG
,
Sandkühler
J
:
Long-term potentiation in spinal nociceptive pathways as a novel target for pain therapy.
Mol Pain
2011
;
7
:
20
225.
Zhang
HM
,
Zhou
LJ
,
Hu
XD
,
Hu
NW
,
Zhang
T
,
Liu
XG
:
Acute nerve injury induces long-term potentiation of C-fiber evoked field potentials in spinal dorsal horn of intact rat.
Sheng Li Xue Bao
2004
;
56
:
591
6
226.
Xu
ZZ
,
Liu
XJ
,
Berta
T
,
Park
CK
,
Lu
N
,
Serhan
CN
,
Ji
RR
:
Neuroprotectin/protectin D1 protects neuropathic pain in mice after nerve trauma.
Ann Neurol
2013
;
74
:
490
5
227.
Drdla
R
,
Gassner
M
,
Gingl
E
,
Sandkühler
J
:
Induction of synaptic long-term potentiation after opioid withdrawal.
Science
2009
;
325
:
207
10
228.
Bonin
RP
,
De Koninck
Y
:
A spinal analog of memory reconsolidation enables reversal of hyperalgesia.
Nat Neurosci
2014
;
17
:
1043
5
229.
Liu
YL
,
Zhou
LJ
,
Hu
NW
,
Xu
JT
,
Wu
CY
,
Zhang
T
,
Li
YY
,
Liu
XG
:
Tumor necrosis factor-α induces long-term potentiation of C-fiber evoked field potentials in spinal dorsal horn in rats with nerve injury: The role of NF-κB, JNK and p38 MAPK.
Neuropharmacology
2007
;
52
:
708
15
230.
Gruber-Schoffnegger
D
,
Drdla-Schutting
R
,
Hönigsperger
C
,
Wunderbaldinger
G
,
Gassner
M
,
Sandkühler
J
:
Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-α and IL-1β is mediated by glial cells.
J Neurosci
2013
;
33
:
6540
51
231.
Chirila
AM
,
Brown
TE
,
Bishop
RA
,
Bellono
NW
,
Pucci
FG
,
Kauer
JA
:
Long-term potentiation of glycinergic synapses triggered by interleukin 1β.
Proc Natl Acad Sci USA
2014
;
111
:
8263
8
232.
Clark
AK
,
Gruber-Schoffnegger
D
,
Drdla-Schutting
R
,
Gerhold
KJ
,
Malcangio
M
,
Sandkühler
J
:
Selective activation of microglia facilitates synaptic strength.
J Neurosci
2015
;
35
:
4552
70
233.
Kronschläger
MT
,
Drdla-Schutting
R
,
Gassner
M
,
Honsek
SD
,
Teuchmann
HL
,
Sandkühler
J
:
Gliogenic LTP spreads widely in nociceptive pathways.
Science
2016
;
354
:
1144
8
234.
Bliss
TV
,
Collingridge
GL
,
Kaang
BK
,
Zhuo
M
:
Synaptic plasticity in the anterior cingulate cortex in acute and chronic pain.
Nat Rev Neurosci
2016
;
17
:
485
96
235.
Bird
GC
,
Lash
LL
,
Han
JS
,
Zou
X
,
Willis
WD
,
Neugebauer
V
:
Protein kinase A-dependent enhanced NMDA receptor function in pain-related synaptic plasticity in rat amygdala neurones.
J Physiol
2005
;
564
:
907
21
236.
Liu
Y
,
Zhou
LJ
,
Wang
J
,
Li
D
,
Ren
WJ
,
Peng
J
,
Wei
X
,
Xu
T
,
Xin
WJ
,
Pang
RP
,
Li
YY
,
Qin
ZH
,
Murugan
M
,
Mattson
MP
,
Wu
LJ
,
Liu
XG
:
TNF-α differentially regulates synaptic plasticity in the hippocampus and spinal cord by microglia-dependent mechanisms after peripheral nerve injury.
J Neurosci
2017
;
37
:
871
81
237.
Sweitzer
SM
,
Schubert
P
,
DeLeo
JA
:
Propentofylline, a glial modulating agent, exhibits antiallodynic properties in a rat model of neuropathic pain.
J Pharmacol Exp Ther
2001
;
297
:
1210
7
238.
Raghavendra
V
,
Tanga
F
,
Rutkowski
MD
,
DeLeo
JA
:
Anti-hyperalgesic and morphine-sparing actions of propentofylline following peripheral nerve injury in rats: Mechanistic implications of spinal glia and proinflammatory cytokines.
Pain
2003
;
104
:
655
64
239.
Landry
RP
,
Jacobs
VL
,
Romero-Sandoval
EA
,
DeLeo
JA
:
Propentofylline, a CNS glial modulator does not decrease pain in post-herpetic neuralgia patients: In vitro evidence for differential responses in human and rodent microglia and macrophages.
Exp Neurol
2012
;
234
:
340
50
240.
Anand
P
,
Shenoy
R
,
Palmer
JE
,
Baines
AJ
,
Lai
RY
,
Robertson
J
,
Bird
N
,
Ostenfeld
T
,
Chizh
BA
:
Clinical trial of the p38 MAP kinase inhibitor dilmapimod in neuropathic pain following nerve injury.
Eur J Pain
2011
;
15
:
1040
8
241.
Ostenfeld
T
,
Krishen
A
,
Lai
RY
,
Bullman
J
,
Baines
AJ
,
Green
J
,
Anand
P
,
Kelly
M
:
Analgesic efficacy and safety of the novel p38 MAP kinase inhibitor, losmapimod, in patients with neuropathic pain following peripheral nerve injury: A double-blind, placebo-controlled study.
Eur J Pain
2013
;
17
:
844
57
242.
Tong
SE
,
Daniels
SE
,
Black
P
,
Chang
S
,
Protter
A
,
Desjardins
PJ
:
Novel p38α mitogen-activated protein kinase inhibitor shows analgesic efficacy in acute postsurgical dental pain.
J Clin Pharmacol
2012
;
52
:
717
28
243.
Sainoh
T
,
Orita
S
,
Miyagi
M
,
Inoue
G
,
Kamoda
H
,
Ishikawa
T
,
Yamauchi
K
,
Suzuki
M
,
Sakuma
Y
,
Kubota
G
,
Oikawa
Y
,
Inage
K
,
Sato
J
,
Nakata
Y
,
Nakamura
J
,
Aoki
Y
,
Toyone
T
,
Takahashi
K
,
Ohtori
S
:
Single intradiscal administration of the tumor necrosis factor-α inhibitor, etanercept, for patients with discogenic low back pain.
Pain Med
2016
;
17
:
40
5
244.
Freeman
BJ
,
Ludbrook
GL
,
Hall
S
,
Cousins
M
,
Mitchell
B
,
Jaros
M
,
Wyand
M
,
Gorman
JR
:
Randomized, double-blind, placebo-controlled, trial of transforaminal epidural etanercept for the treatment of symptomatic lumbar disc herniation.
Spine (Phila Pa 1976)
2013
;
38
:
1986
94
245.
Terkeltaub
R
,
Sundy
JS
,
Schumacher
HR
,
Murphy
F
,
Bookbinder
S
,
Biedermann
S
,
Wu
R
,
Mellis
S
,
Radin
A
:
The interleukin 1 inhibitor rilonacept in treatment of chronic gouty arthritis: Results of a placebo-controlled, monosequence crossover, non-randomised, single-blind pilot study.
Ann Rheum Dis
2009
;
68
:
1613
7
246.
Roerink
ME
,
Bredie
SJH
,
Heijnen
M
,
Dinarello
CA
,
Knoop
H
,
Van der Meer
JWM
:
Cytokine inhibition in patients with chronic fatigue syndrome: A randomized trial.
Ann Intern Med
2017
;
166
:
557
64
247.
Wang
X
,
Zhang
Y
,
Peng
Y
,
Hutchinson
MR
,
Rice
KC
,
Yin
H
,
Watkins
LR
:
Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4.
Br J Pharmacol
2016
;
173
:
856
69
248.
Younger
J
,
Mackey
S
:
Fibromyalgia symptoms are reduced by low-dose naltrexone: A pilot study.
Pain Med
2009
;
10
:
663
72
249.
Echeverry
S
,
Shi
XQ
,
Yang
M
,
Huang
H
,
Wu
Y
,
Lorenzo
LE
,
Perez-Sanchez
J
,
Bonin
RP
,
De Koninck
Y
,
Zhang
J
:
Spinal microglia are required for long-term maintenance of neuropathic pain.
Pain
2017
;
158
:
1792
801
250.
Frampton
M
,
Harvey
RJ
,
Kirchner
V
:
Propentofylline for dementia.
Cochrane Database Syst Rev
2003
, pp
CD002853
251.
Yekkirala
AS
,
Roberson
DP
,
Bean
BP
,
Woolf
CJ
:
Breaking barriers to novel analgesic drug development.
Nat Rev Drug Discov
2017
;
16
:
545
64
252.
Mogil
JS
:
Animal models of pain: Progress and challenges.
Nat Rev Neurosci
2009
;
10
:
283
94
253.
King
T
,
Vera-Portocarrero
L
,
Gutierrez
T
,
Vanderah
TW
,
Dussor
G
,
Lai
J
,
Fields
HL
,
Porreca
F
:
Unmasking the tonic-aversive state in neuropathic pain.
Nat Neurosci
2009
;
12
:
1364
6
254.
Langford
DJ
,
Bailey
AL
,
Chanda
ML
,
Clarke
SE
,
Drummond
TE
,
Echols
S
,
Glick
S
,
Ingrao
J
,
Klassen-Ross
T
,
Lacroix-Fralish
ML
,
Matsumiya
L
,
Sorge
RE
,
Sotocinal
SG
,
Tabaka
JM
,
Wong
D
,
van den Maagdenberg
AM
,
Ferrari
MD
,
Craig
KD
,
Mogil
JS
:
Coding of facial expressions of pain in the laboratory mouse.
Nat Methods
2010
;
7
:
447
9
255.
Rosenberg
MB
,
Carroll
FI
,
Negus
SS
:
Effects of monoamine reuptake inhibitors in assays of acute pain-stimulated and pain-depressed behavior in rats.
J Pain
2013
;
14
:
246
59
256.
Cobos
EJ
,
Ghasemlou
N
,
Araldi
D
,
Segal
D
,
Duong
K
,
Woolf
CJ
:
Inflammation-induced decrease in voluntary wheel running in mice: A nonreflexive test for evaluating inflammatory pain and analgesia.
Pain
2012
;
153
:
876
84
257.
Serhan
CN
,
Chiang
N
,
Van Dyke
TE
:
Resolving inflammation: Dual anti-inflammatory and pro-resolution lipid mediators.
Nat Rev Immunol
2008
;
8
:
349
61
258.
Ji
RR
,
Xu
ZZ
,
Strichartz
G
,
Serhan
CN
:
Emerging roles of resolvins in the resolution of inflammation and pain.
Trends Neurosci
2011
;
34
:
599
609
259.
Huang
L
,
Wang
CF
,
Serhan
CN
,
Strichartz
G
:
Enduring prevention and transient reduction of postoperative pain by intrathecal resolvin D1.
Pain
2011
;
152
:
557
65
260.
Serhan
CN
,
Dalli
J
,
Karamnov
S
,
Choi
A
,
Park
CK
,
Xu
ZZ
,
Ji
RR
,
Zhu
M
,
Petasis
NA
:
Macrophage proresolving mediator maresin 1 stimulates tissue regeneration and controls pain.
FASEB J
2012
;
26
:
1755
65
261.
Park
CK
,
Xu
ZZ
,
Liu
T
,
N
,
Serhan
CN
,
Ji
RR
:
Resolvin D2 is a potent endogenous inhibitor for transient receptor potential subtype V1/A1, inflammatory pain, and spinal cord synaptic plasticity in mice: Distinct roles of resolvin D1, D2, and E1.
J Neurosci
2011
;
31
:
18433
8
262.
Svensson
CI
,
Zattoni
M
,
Serhan
CN
:
Lipoxins and aspirin-triggered lipoxin inhibit inflammatory pain processing.
J Exp Med
2007
;
204
:
245
52
263.
Wang
JC
,
Strichartz
GR
:
Prevention of chronic post-thoracotomy pain in rats by intrathecal resolvin D1 and D2: Effectiveness of perioperative and delayed drug delivery.
J Pain
2017
;
18
:
535
45
264.
Terrando
N
,
Gómez-Galán
M
,
Yang
T
,
Carlström
M
,
Gustavsson
D
,
Harding
RE
,
Lindskog
M
,
Eriksson
LI
:
Aspirin-triggered resolvin D1 prevents surgery-induced cognitive decline.
FASEB J
2013
;
27
:
3564
71
265.
Kehlet
H
,
Jensen
TS
,
Woolf
CJ
:
Persistent postsurgical pain: Risk factors and prevention.
Lancet
2006
;
367
:
1618
25
266.
Chen
G
,
Park
CK
,
Xie
RG
,
Ji
RR
:
Intrathecal bone marrow stromal cells inhibit neuropathic pain via TGF-β secretion.
J Clin Invest
2015
;
125
:
3226
40
267.
Guo
W
,
Wang
H
,
Zou
S
,
Gu
M
,
Watanabe
M
,
Wei
F
,
Dubner
R
,
Huang
GT
,
Ren
K
:
Bone marrow stromal cells produce long-term pain relief in rat models of persistent pain.
Stem Cells
2011
;
29
:
1294
303
268.
Bráz
JM
,
Sharif-Naeini
R
,
Vogt
D
,
Kriegstein
A
,
Alvarez-Buylla
A
,
Rubenstein
JL
,
Basbaum
AI
:
Forebrain GABAergic neuron precursors integrate into adult spinal cord and reduce injury-induced neuropathic pain.
Neuron
2012
;
74
:
663
75
269.
Lee
MJ
,
Yoon
TG
,
Kang
M
,
Kim
HJ
,
Kang
KS
:
Effect of subcutaneous treatment with human umbilical cord blood-derived multipotent stem cells on peripheral neuropathic pain in rats.
Korean J Physiol Pharmacol
2017
;
21
:
153
60
270.
Willemen
HL
,
Eijkelkamp
N
,
Garza Carbajal
A
,
Wang
H
,
Mack
M
,
Zijlstra
J
,
Heijnen
CJ
,
Kavelaars
A
:
Monocytes/macrophages control resolution of transient inflammatory pain.
J Pain
2014
;
15
:
496
506
271.
Krukowski
K
,
Eijkelkamp
N
,
Laumet
G
,
Hack
CE
,
Li
Y
,
Dougherty
PM
,
Heijnen
CJ
,
Kavelaars
A
:
CD8+ T cells and endogenous IL-10 are required for resolution of chemotherapy-induced neuropathic pain.
J Neurosci
2016
;
36
:
11074
83
272.
Barreto
A
,
Braun
TR
:
A method to induce interleukin-1 receptor antagonist protein from autologous whole blood.
Cytokine
2016
;
81
:
137
41
273.
Meijer
H
,
Reinecke
J
,
Becker
C
,
Tholen
G
,
Wehling
P
:
The production of anti-inflammatory cytokines in whole blood by physico-chemical induction.
Inflamm Res
2003
;
52
:
404
7
274.
Shen
L
,
Yuan
T
,
Chen
S
,
Xie
X
,
Zhang
C
:
The temporal effect of platelet-rich plasma on pain and physical function in the treatment of knee osteoarthritis: Systematic review and meta-analysis of randomized controlled trials.
J Orthop Surg Res
2017
;
12
:
16
275.
Moreno-Duarte
I
,
Morse
LR
,
Alam
M
,
Bikson
M
,
Zafonte
R
,
Fregni
F
:
Targeted therapies using electrical and magnetic neural stimulation for the treatment of chronic pain in spinal cord injury.
Neuroimage
2014
;
85
:
1003
13
276.
Shamji
MF
,
De Vos
C
,
Sharan
A
:
The advancing role of neuromodulation for the management of chronic treatment-refractory pain.
Neurosurgery
2017
;
80
:
108
13
277.
Pawela
CP
,
Kramer
JM
,
Hogan
QH
:
Dorsal root ganglion stimulation attenuates the BOLD signal response to noxious sensory input in specific brain regions: Insights into a possible mechanism for analgesia.
Neuroimage
2017
;
147
:
10
8
278.
Ji
RR
,
Schlaepfer
TE
,
Aizenman
CD
,
Epstein
CM
,
Qiu
D
,
Huang
JC
,
Rupp
F
:
Repetitive transcranial magnetic stimulation activates specific regions in rat brain.
Proc Natl Acad Sci USA
1998
;
95
:
15635
40
279.
Yang
F
,
Zhang
T
,
Tiwari
V
,
Shu
B
,
Zhang
C
,
Wang
Y
,
Vera-Portocarrero
LP
,
Raja
SN
,
Guan
Y
:
Effects of combined electrical stimulation of the dorsal column and dorsal roots on wide-dynamic-range neuronal activity in nerve-injured rats.
Neuromodulation
2015
;
18
:
592
7
;
discussion 598
280.
Zhang
TC
,
Janik
JJ
,
Peters
RV
,
Chen
G
,
Ji
RR
,
Grill
WM
:
Spinal sensory projection neuron responses to spinal cord stimulation are mediated by circuits beyond gate control.
J Neurophysiol
2015
;
114
:
284
300
281.
Han
JS
:
Acupuncture: Neuropeptide release produced by electrical stimulation of different frequencies.
Trends Neurosci
2003
;
26
:
17
22
282.
Borovikova
LV
,
Ivanova
S
,
Zhang
M
,
Yang
H
,
Botchkina
GI
,
Watkins
LR
,
Wang
H
,
Abumrad
N
,
Eaton
JW
,
Tracey
KJ
:
Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin.
Nature
2000
;
405
:
458
62
283.
Steinberg
BE
,
Sundman
E
,
Terrando
N
,
Eriksson
LI
,
Olofsson
PS
:
Neural control of inflammation: Implications for perioperative and critical care.
Anesthesiology
2016
;
124
:
1174
89
284.
Goldman
N
,
Chen
M
,
Fujita
T
,
Xu
Q
,
Peng
W
,
Liu
W
,
Jensen
TK
,
Pei
Y
,
Wang
F
,
Han
X
,
Chen
JF
,
Schnermann
J
,
Takano
T
,
Bekar
L
,
Tieu
K
,
Nedergaard
M
:
Adenosine A1 receptors mediate local anti-nociceptive effects of acupuncture.
Nat Neurosci
2010
;
13
:
883
8
285.
Torres-Rosas
R
,
Yehia
G
,
Peña
G
,
Mishra
P
,
del Rocio Thompson-Bonilla
M
,
Moreno-Eutimio
MA
,
Arriaga-Pizano
LA
,
Isibasi
A
,
Ulloa
L
:
Dopamine mediates vagal modulation of the immune system by electroacupuncture.
Nat Med
2014
;
20
:
291
5
286.
Maixner
W
,
Randich
A
:
Role of the right vagal nerve trunk in antinociception.
Brain Res
1984
;
298
:
374
7
287.
Fang
JF
,
Du
JY
,
Shao
XM
,
Fang
JQ
,
Liu
Z
:
Effect of electroacupuncture on the NTS is modulated primarily by acupuncture point selection and stimulation frequency in normal rats.
BMC Complement Altern Med
2017
;
17
:
182
288.
Lim
HD
,
Kim
MH
,
Lee
CY
,
Namgung
U
:
Anti-inflammatory effects of acupuncture stimulation via the vagus nerve.
PLoS One
2016
;
11
:
e0151882
289.
da Silva
MD
,
Bobinski
F
,
Sato
KL
,
Kolker
SJ
,
Sluka
KA
,
Santos
AR
:
IL-10 cytokine released from M2 macrophages is crucial for analgesic and anti-inflammatory effects of acupuncture in a model of inflammatory muscle pain.
Mol Neurobiol
2015
;
51
:
19
31