Abstract

Objective This article reviews the structural and functional changes in pain chronification and explores the association between memory and the development of chronic pain.

Methods PubMed was searched using the terms “chronic pain,” “central sensitization,” “learning,” “memory,” “long-term potentiation,” “long-term depression,” and “pain memory.” Relevant findings were synthesized into a narrative of the processes affecting pain chronification.

Results Pain pathways represent a complex sensory system with cognitive, emotional, and behavioral influences. Anatomically, the hippocampus, amygdala, and anterior cortex—central to the encoding and consolidation of memory—are also implicated in experiential aspects of pain. Common neurotransmitters and similar mechanisms of neural plasticity (eg, central sensitization, long-term potentiation) suggest a mechanistic overlap between chronic pain and memory. These anatomic and mechanistic correlates indicate that chronic pain and memory intimately interact on several levels. Longitudinal imaging studies suggest that spatiotemporal reorganization of brain activity accompanies the transition to chronic pain, during which the representation of pain gradually shifts from sensory to emotional and limbic structures.

Conclusions The chronification of pain can be conceptualized as activity-induced plasticity of the limbic–cortical circuitry resulting in reorganization of the neocortex. The state of the limbic–cortical network determines whether nociceptive signals are transient or chronic by extinguishing pathways or amplifying signals that intensify the emotional component of nociceptive inputs. Thus, chronic pain can be seen as the persistence of the memory of pain and/or the inability to extinguish painful memories. Ideally, pharmacologic, physical, and/or psychological approaches should reverse the reorganization accompanying chronic pain.

Introduction

Chronic pain is one of the primary reasons patients seek health care in the United States, with a significant proportion of physician visits attributable to some type of chronic pain complaint [1]. The International Association for the Study of Pain (IASP) provides a definition of chronic pain that accounts for both the duration and complexity of pain (ie, persistent or recurrent pain lasting longer than three months) [2]. In addition, the IASP, in cooperation with the World Health Organization, has proposed a rational classification for chronic pain based on etiology, underlying pathophysiological mechanisms, and body site [2].

On a community level, the prevalence and cost burden of chronic pain are considerable, and the individual distress caused can also be substantial, with cognitive impairment, depression, and loss of function [3,4]. According to an Institute of Medicine US report, chronic pain affects more than 100 million American adults—more than those affected by heart disease, cancer, and diabetes combined—and costs up to $635 billion each year in medical treatment and lost productivity [5].

Traditionally, chronic pain has been categorized as chronic cancer pain and chronic noncancer pain. However, this distinction is largely philosophical, with little, if any, medical or clinical relevance [6], and it does not help clinicians to better understand the mechanisms underlying pain chronification or provide guidance for appropriate treatment approaches. Understanding the mechanisms that underlie chronic pain and the processes associated with its development can help inform treatment. Thus, the primary goal of this article is to explore the association between learning, memory, and the development and perpetuation of chronic pain. Because the topics covered in this review are complex, this article begins with a brief overview of the transmission of pain in the peripheral and central nervous system (CNS) and the structural and functional changes in pain chronification, which are important for understanding how these processes relate to learning and memory.

Pain Pathways and Mechanisms: From Nociception to Central Processing

Pain pathways represent a complex sensory system, with cognitive, emotional, and behavioral elements having evolved to detect and integrate a protective response to noxious stimuli [7]. In humans, this system involves both primitive spinal reflexes and complex conscious and subconscious supraspinal responses. The intensity and spatial and temporal patterns of noxious stimuli are transduced into a signal through nociceptors and carried by thinly myelinated or unmyelinated nerve fibers (Aδ and C fibers, respectively), which are then conveyed to the CNS and ultimately to higher CNS centers (vide infra) (Figure 1 ) [9–11]. Along with other touch sensory neurons, the cell bodies of primary afferent Aδ and C fibers are located in the dorsal root ganglion [8,12] and synapse onto several cell types in the spinal dorsal horn, releasing excitatory neurotransmitters including glutamate, substance P, calcitonin gene–related peptide (CGRP), and other peptides [9,10]. In addition to synapsing with interneurons involved in local reflexes, primary afferents synapse onto nociceptive-specific neurons involved in signaling the presence and location of pain and inhibitory and excitatory interneurons in the Rexed lamina I and II (substantia gelatinosa) [9,12]. Primary afferent neurons also synapse onto projection neurons located more deeply in the dorsal horn, termed “wide dynamic range” neurons, which signal the intensity of pain, and their response characteristics can be modified by inputs [13–15].

Figure 1

Ascending and descending pain pathways (adapted from Zhuo [8]). ACC = anterior cingulate cortex; DRG = dorsal root ganglion; GABA = gamma aminobutyric acid; Glu = glutamate; HC = hippocampus; IC = insular cortex; NRM = nucleus raphe magnus; PAG = periaqueductal gray; PFC = prefrontal cortex; S1 = primary somatosensory cortex; S2 = secondary somatosensory cortex; SDH = spinal dorsal horn; VPL = ventral posterolateral nucleus of the thalamus.

Projection neurons from the dorsal horn decussate at the ventral commissure, ascend in the lateral spinothalamic tract, and end in the ventral posterolateral nuclei of the thalamus, with relays to the somatosensory cortex and periaqueductal gray matter (PAG) [9,12]. Ascending spinoreticular and spinomesencephalic tracts carry pain and touch information to areas that play a role in the memory and affective components of pain such as the amygdala, hypothalamus, and PAG [9,16].

Supraspinal involvement is fundamental to the experiential aspects of pain. Multiple regions are involved, including the hypothalamus, amygdala, and cortex—specifically, the anterior cingulate cortex, insular cortex, primary cortex, and secondary somatosensory cortex—as well as the nucleus accumbens (NAc) and the PAG (Figure 1) [8,9,12,17–21].

A larger system involves the function of various subsystems, including the descending pain modulatory system encompassing the PAG and rostral ventromedial medulla (RVM), as well as the reward-motivation network, including the NAc, the prefrontal cortex, and the ventral tegmental area [9,18,19]. These descending tracts are monoaminergic, and their activation from the RVM and PAG can release norepinephrine and serotonin into the spinal dorsal horn [22,23].

Glial cells also play a role in the release of neurotransmitters and molecules involved in pain processing (eg, CGRP, substance P, and glutamate) and are critically important in the processing, expression, and transmission of pain, as well as in the development of chronic pain [24,25]. In addition, glia can release inflammatory cytokines known to be involved in pain [26–29]. The multifaceted interaction between glia and neurons depends on glial cell types, the location of the regulatory process (peripheral nerve, spinal cord, or brain), and the type of the pain (eg, acute, chronic) [30,31].

The concept of simple transmission of pain signals in the spinal cord fails to account for the complex interactions that occur in the dorsal horn and elsewhere. Central sensitization of dorsal horn neurons constitutes a form of long-term plasticity in the CNS and often occurs following injury and inflammation. It is characterized by increased excitability of dorsal horn neurons, an increase in spontaneous activity, enlarged receptive field areas, and an increase in responses evoked by large- and small-caliber primary afferent fibers (Figure 2 ) [32–35]. The mechanism underlying central sensitization involves glutamate released by C fibers onto postsynaptic N-methyl-D-aspartate (NMDA) receptors, causing calcium influx [36,37]. The release of glutamate and substance P is mediated through presynaptic neuronal N-type voltage-gated calcium channels [38]. In addition to sensitization of the dorsal horn, calcium flux through NMDA receptors is critical to synaptic plasticity [33]. Indeed, central sensitization involves changes in the spinal cord gray matter, which mimic the supraspinal plasticity that accompanies both learning and memory [39], which is discussed further in the next section.

Figure 2

Central sensitization in the dorsal horn of the spinal cord (adapted from Kuner [32]). Nociceptor afferent may increase the strength of synapses between these fibers and dorsal horn neurons. These changes can result in stronger responses to a given stimulus (sensitization) and expansion of the area on the body surface to which a given dorsal horn cell responds (ie, its receptive field).

“Windup”—which refers to progressive increases in the magnitude of C-fiber-evoked responses during a continuous stimulus in experimental settings—shares some features with central sensitization and provides a theoretical foundation for understanding the role of neural plasticity in the development of chronic pain [40,41]. Although the analysis of the cellular and molecular mechanisms underlying central sensitization and synaptic plasticity that accompanies learning and memory suggests some subtle differences, there are several mechanistic similarities, including similar pharmacology (ie, specific glutamate receptors and molecules involved in calcium signaling) and structural changes, which are further discussed in the next section.

Anatomic and Physiologic Overlap with Pain, Memory, and Learning

Considerable progress has been made toward understanding the neurologic underpinnings of memory, learning, and chronic pain, thereby permitting greater insight into common anatomical systems, and neurochemical substrates.

Memory is defined as explicit (declarative; eg, remembering a list of dates) or implicit (nondeclarative; eg, tying shoelaces) [42]. Implicit memory includes associative and nonassociative learning [43]. Encoding is a critical initial step in memory formation that is initiated by attention to an event (regulated by the thalamus and frontal lobe) causing more frequent neuronal firing [12,42,44,45]. Attention is increased by emotional content and processed on an unconscious level in the amygdala. These sensations are then translated experientially in sensory areas of the cortex before being combined in the hippocampus. The hippocampus and amygdala interact synergistically to form long-term memories of significantly emotional events [46,47], and the amygdala is a key site of synaptic plasticity associated with fear conditioning (a form of associative learning) [48].

Explicit and implicit forms of memory are distinct from pain memory—the neuroplasticity leading to memory-like maladaptive changes in response to painful stimuli [49]. Nevertheless, neuroplasticity is one of the neurochemical bases of learning and memory, as well as the development and persistence of chronic pain (Table 1, Figure 3) [33,50]. This involves long-term potentiation (LTP), which refers to a persistent increase in the efficacy of synaptic transmission when the same neurons are active at the same time and become sensitized to each other [33,45,50]. Windup is an enduring form of synaptic plasticity that shares essential biochemical features with some forms of LTP associated with memory consolidation, including the activation of NMDA receptors [37,50,51,68]. Central sensitization in the spinal cord is characterized by alterations in glutamate transmission through NMDA receptors and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, similar to changes underlying LTP in learning and memory [33,48,52,69].

Table 1

Anatomic and physiologic overlap between pain, learning, and memory [9,33,46–48,50–67]

Shared FeaturesChronic/Persistent PainLearning and Memory
Synaptic plasticityActivity-dependent synaptic plasticity (wind-up, central sensitization) underlies inflammatory and neuropathic painLearning and memory rely on activity-dependent LTP at CNS synapses
NMDA receptor involvement in central sensitizationNMDA receptor activation involvement in LTP
BDNF enhances C-fiber-evoked responses in central sensitizationBDNF contributes to synaptic plasticity associated with various types of learning
Serotonin dysregulation is a feature of chronic painSerotonin signaling plays a role in learning and memory through interactions with other neurotransmitter systems
Calcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to central sensitizationCalcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to LTP associated with memory
Structural changesCentral sensitization involves protein synthesis and synaptogenesisLTP involves protein synthesis and synaptogenesis
Changes in connectivity in limbic and cortical areas contribute to chronic painCortical reorganization accompanies learning and memory
Anatomical overlapCortico-limbic pathways are involved in transition to pain chronicityCortico-limbic circuits are crucial for emotional learning
Chronic pain is associated with functional changes in the hippocampus and amygdalaHippocampus and amygdala act synergistically in the formation of long-term memories of significantly emotional events
Hippocampal activity is associated with exacerbation of pain by anxiety
Shared FeaturesChronic/Persistent PainLearning and Memory
Synaptic plasticityActivity-dependent synaptic plasticity (wind-up, central sensitization) underlies inflammatory and neuropathic painLearning and memory rely on activity-dependent LTP at CNS synapses
NMDA receptor involvement in central sensitizationNMDA receptor activation involvement in LTP
BDNF enhances C-fiber-evoked responses in central sensitizationBDNF contributes to synaptic plasticity associated with various types of learning
Serotonin dysregulation is a feature of chronic painSerotonin signaling plays a role in learning and memory through interactions with other neurotransmitter systems
Calcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to central sensitizationCalcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to LTP associated with memory
Structural changesCentral sensitization involves protein synthesis and synaptogenesisLTP involves protein synthesis and synaptogenesis
Changes in connectivity in limbic and cortical areas contribute to chronic painCortical reorganization accompanies learning and memory
Anatomical overlapCortico-limbic pathways are involved in transition to pain chronicityCortico-limbic circuits are crucial for emotional learning
Chronic pain is associated with functional changes in the hippocampus and amygdalaHippocampus and amygdala act synergistically in the formation of long-term memories of significantly emotional events
Hippocampal activity is associated with exacerbation of pain by anxiety

BDNF = brain-derived neurotrophic factor; CNS = central nervous system; LTP = long-term potentiation; NMDA = N-methyl-D-aspartate.

Table 1

Anatomic and physiologic overlap between pain, learning, and memory [9,33,46–48,50–67]

Shared FeaturesChronic/Persistent PainLearning and Memory
Synaptic plasticityActivity-dependent synaptic plasticity (wind-up, central sensitization) underlies inflammatory and neuropathic painLearning and memory rely on activity-dependent LTP at CNS synapses
NMDA receptor involvement in central sensitizationNMDA receptor activation involvement in LTP
BDNF enhances C-fiber-evoked responses in central sensitizationBDNF contributes to synaptic plasticity associated with various types of learning
Serotonin dysregulation is a feature of chronic painSerotonin signaling plays a role in learning and memory through interactions with other neurotransmitter systems
Calcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to central sensitizationCalcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to LTP associated with memory
Structural changesCentral sensitization involves protein synthesis and synaptogenesisLTP involves protein synthesis and synaptogenesis
Changes in connectivity in limbic and cortical areas contribute to chronic painCortical reorganization accompanies learning and memory
Anatomical overlapCortico-limbic pathways are involved in transition to pain chronicityCortico-limbic circuits are crucial for emotional learning
Chronic pain is associated with functional changes in the hippocampus and amygdalaHippocampus and amygdala act synergistically in the formation of long-term memories of significantly emotional events
Hippocampal activity is associated with exacerbation of pain by anxiety
Shared FeaturesChronic/Persistent PainLearning and Memory
Synaptic plasticityActivity-dependent synaptic plasticity (wind-up, central sensitization) underlies inflammatory and neuropathic painLearning and memory rely on activity-dependent LTP at CNS synapses
NMDA receptor involvement in central sensitizationNMDA receptor activation involvement in LTP
BDNF enhances C-fiber-evoked responses in central sensitizationBDNF contributes to synaptic plasticity associated with various types of learning
Serotonin dysregulation is a feature of chronic painSerotonin signaling plays a role in learning and memory through interactions with other neurotransmitter systems
Calcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to central sensitizationCalcium signaling through NMDA receptors or voltage-dependent calcium channels contributes to LTP associated with memory
Structural changesCentral sensitization involves protein synthesis and synaptogenesisLTP involves protein synthesis and synaptogenesis
Changes in connectivity in limbic and cortical areas contribute to chronic painCortical reorganization accompanies learning and memory
Anatomical overlapCortico-limbic pathways are involved in transition to pain chronicityCortico-limbic circuits are crucial for emotional learning
Chronic pain is associated with functional changes in the hippocampus and amygdalaHippocampus and amygdala act synergistically in the formation of long-term memories of significantly emotional events
Hippocampal activity is associated with exacerbation of pain by anxiety

BDNF = brain-derived neurotrophic factor; CNS = central nervous system; LTP = long-term potentiation; NMDA = N-methyl-D-aspartate.

Figure 3

Long-term potentiation (adapted from Lamprecht and LeDoux [50]). As in central sensitization, specific stimulation results in strengthening of synapses that may involve multiple mechanisms. This strengthening results in a greater response of the postsynaptic cell to a given presynaptic input. LTP = long-term potentiation.

Animal studies suggest that while LTP plays a role in encoding memory, long-term depression (LTD; persistent reduction in synaptic strength) can inactivate memory [70]. Similarly, LTD has been observed in nociceptive pathways and is associated with reduced pain sensitivity or hyperalgesia [49,71].

The transience of short-term memory reflects the transitory nature of underlying biochemical changes, whereas long-term memory involves new neuronal processes and synapses arising from alterations in protein synthesis and gene regulation [42,45]. Indeed, synaptogenesis plays a central role in learning and memory [53,72], and this process is important in the development of chronic pain [54]. Long-term potentiation represents post-translational plasticity, which persists when it is followed by gene transcription, protein translation, and protein synthesis, which act to consolidate changes at synapses [73]. In memory consolidation, additional connections and pathways are created, with new experiences leading to rerouting of connections and reorganization [55], reinforced by the synthesis of new proteins to remodel synapses [74]. In this manner, the brain undergoes reorganization in response to experiences, creating new memories that are triggered and shaped by experience. Eventually, memory is stored in other areas of the brain, including the striatum (memories for skills and habits) [75,76], the neocortex (priming), and the cerebellum (simple forms of associative learning) [77,78].

Although neurons have traditionally been the focus of neuroplasticity in memory and learning, glia also play a critical role in regulating synaptic and structural plasticity in areas involved in not only learning and memory, but also pain [30,31,79–81]. Oligodendrocytes also support the production of myelin [82], which is essential to the fast impulse conduction tracts required for memory formation [83,84]. Thus, the involvement of glia represents another functional similarity between the plasticity underlying learning and memory and chronic pain.

Chronic pain and learning and memory share anatomical sites of synaptic plasticity or functional changes (Table 1). For example, in addition to their roles in learning and memory, the hippocampus and/or amygdala also likely play a role in chronic pain and are subject to pain-induced plasticity. In murine models, chronic pain has been associated with alterations in hippocampus-dependent behavior, and cellular and molecular fluctuations in the hippocampus and/or amygdala—including changes in volume, neurogenesis, and altered synaptic plasticity—have been detected [56–60]. Chronic pain–related changes have been observed in the anterior cingulate cortex and NAc as well [85–87]. Clinical experience suggests an anatomical overlap between chronic pain and learning and memory [61,88]. It has been recognized for some time that pain is exacerbated by anxiety, and individuals differ in their sensitivity to pain expectancy. Specifically, activity in the hippocampal network is associated with exacerbation of pain by anxiety, whereas individual sensitivity to pain expectancy is related to differential activation of the hippocampus and amygdala [61,88].

Another mechanistic similarity between chronic pain and learning and memory is the involvement of neuronal growth factors in synaptic plasticity. For example, the persistent synaptic plasticity underlying the formation of object–place and spatial reference memory in mice requires brain-derived neurotrophic factor (BDNF) [89]. Similarly, spinal BDNF signaling contributes to the synaptic plasticity underlying neuropathic pain in a rat model of nerve injury, and supraspinal BDNF facilitates pain in a model of persistent inflammatory pain [90].

On a neurochemical level, the presence of serotonin markers in the brain areas involved in memory and learning is well documented [91–96]. Serotonin is projected widely throughout the brain [97], and a large body of evidence indicates the interplay between serotonergic transmission and other neurotransmitters (eg, dopamine, glutamate, and gamma-amino butyric acid [GABA]) in the neurobiological control of learning and memory [62,63,98]. Dysregulation of serotonin signaling also plays an important role in chronic pain [9,62].

Mechanistically, serotonin is involved in short-term memory and learning by modulating neuronal membrane channels, including those that directly regulate neurotransmitter release (ie, Ca2+ channels in the presynaptic neuron), neuronal excitability, and synaptic responses in postsynaptic neurons [39,45]. Specifically, serotonin binds to neuron receptors coupled to either the diacylglycerol-protein kinase C system or the cyclic adenosine monophosphate-protein kinase A system [39,45]. Through phosphorylation, these protein kinases regulate the properties of membrane channels and other processes involved in neurotransmitter release. Serotonin also leads to changes in the properties of the postsynaptic motor neuron, including an increase in the number of glutamate receptors [9,62].

Overall, there is mounting evidence of anatomic and physiologic overlap between pain and memory on several levels. Anatomically, preclinical and clinical evidence indicates that several neuroanatomical substrates are common in these processes. The hippocampus and amygdala and anterior cortex are central to the encoding and consolidation of memory, including the emotional content, and these structures are implicated in the experiential aspects of pain. Mechanistic overlap between pain and memory is plausible because several neurotransmitters and the process of LTP are common to both. These anatomic and mechanistic correlates strengthen the hypothesis that these phenomena do not exist in isolation, but instead intimately interact on several levels.

Chronic Pain, Memory, and Learning

Evidence from Neuroimaging Studies

Clinical studies using neuroimaging techniques, including magnetic resonance imaging (MRI), functional MRI (fMRI), and magnetic resonance spectroscopy (MRS), have greatly advanced our understanding of the interaction between pain and memory/learning on neuroanatomical and functional levels. More than a decade ago, neuroimaging demonstrated that cortical gray matter density decreased regionally in chronic back pain [99]. In this early report, patients with chronic back pain had 5% to 11% less neocortical gray matter volume than control subjects—estimated to be equivalent to the gray matter volume lost in 10 to 20 years of normal aging [100,101].

Since this initial finding, mounting clinical evidence has documented brain morphological changes in various chronic pain conditions (eg, back pain, fibromyalgia, complex regional pain syndrome, knee osteoarthritis, irritable bowel syndrome, headache) [102–112]. Using voxel-based morphometry (VBM) and other techniques, conspicuous and varied patterns of altered brain morphology have been observed in a number of chronic pain conditions. For example, gray matter reductions in the amygdala and several cortical areas were observed in patients with chronic migraine vs healthy controls [108], and increases were seen in the striatum and orbitofrontal cortex in patients with fibromyalgia [111]. Of interest, a recent MRI study found significantly less bilateral hippocampal volume compared with controls in patients with some chronic pain conditions [59]. These findings are consistent with the notion that the regions involved in memory, particularly the hippocampus and amygdala, are altered morphologically in patients with chronic pain.

In general, cross-sectional fMRI studies indicate preferential involvement of the limbic and emotional systems in encoding pain when the pain is chronic [64, 113–121]. For example, in patients with trigeminal neuralgia, a recent structural (VBM) and functional (MRS) neuroimaging study showed reductions in gray matter volume compared with controls [119]. This was seen in the thalamus, putamen, primary somatosensory cortex, anterior insular cortex, and NAc. A coincident increase in gray matter volume in the posterior insula was also observed. In the same study, MRS revealed reductions in N-acetylaspartate/creatine ratio (NAA/Cr)—a biochemical marker of neural viability [122]—in the region of thalamic volume loss. The NAA/Cr changes correlated with pain duration and intensity. Results from other studies using various neuroimaging approaches further support the structural and functional plasticity in limbic and cortical sites that accompanies chronic pain [123, 124].

In addition to morphological and functional alterations, chronic pain also appears to modify brain dynamics by altering brain resting state interactions between networks implicated in attention, salience, and reward [125–129]. Together, findings such as these support a mechanistic model for pain chronification in which nociceptive inputs contribute to structural and functional plasticity in the limbic–cortical circuitry. Learning mechanisms within the limbic circuitry may contribute to the transition from acute to chronic pain.

Within the context of this model, two recent longitudinal brain imaging studies have explored the reorganization of brain activity that accompanies and perhaps mediates the transition to chronicity [64,65]. In one study, patients with subacute back pain (SBP), defined as pain lasting four to 16 weeks, were assessed over one year by T1-weighted MRI, VBM, and fMRI as patients transitioned to persistent pain or recovery, thereby allowing comparisons with healthy and chronic back pain subjects [65]. Persistent or recovering patients with SBP were subdivided based on a self-reported 20% change in pain intensity from baseline to one year. Notably, over the study period, patients with persistent SBP exhibited regional brain morphologic changes including decreased gray matter density in the insular cortex, primary somatosensory and motor cortex, and NAc, a key part of the mesolimbic circuitry underlying reinforcement learning. In contrast, across all measures and time points, patients who recovered from SBP resembled healthy controls [65]. Thus, in this study, gray matter density decreased in patients with persistent SBP, but not in those who recovered from SBP.

Related alterations in functional connectivity were also observed in those with persistent pain, which collectively correlated with back pain intensity in the second study. Within the one-year study period, back pain–related brain activity in patients with persistent SBP transitioned from the insular cortex, anterior cingulate cortex, thalamus, and basal ganglia to the medial prefrontal cortex, amygdala, and basal ganglia [64]. Although the transition was slow, it was preceded by functional connectivity differences detectable as soon as the first brain scan [64]. Moreover, the baseline medial prefrontal cortex–NAc functional connectivity strength predicted the development of chronic back pain with 81% accuracy [65], suggesting that enhanced mesolimbic circuitry drives brain reorganization [130].

This temporal profile of brain reorganization during pain chronification and the assumption that early vs chronic stages of back pain are preferentially associated with acute pain or emotion circuitry, respectively, were subsequently tested by the same investigators. Patients with early, acute/SBP (defined as pain lasting approximately two months with no prior history of back pain for one year) were compared with the patients with chronic back pain (defined as pain lasting >10 years). Persistent or recovering SBP patients were defined as in the previous study. Investigators found that brain activity for back pain in patients in the combined early, acute/subacute group was limited to the regions involved in acute pain, whereas in the chronic back pain group, activity was confined to emotion-related circuitry. Among the subset followed longitudinally for one year, patients with recovering and persistent SBP showed activation within acute pain regions at the first two follow-up visits encompassing the bilateral insula, thalamus, and anterior cingulate cortex. However, patients recovering from SBP showed no significant activity for the final two visits, whereas patients with persistent SBP exhibited increased activation in the medial prefrontal cortex and amygdala at the final visit (Figure 4 ) [64]. This suggests that spatiotemporal reorganization of brain activity accompanies the transition to chronic pain, during which the representation of back pain gradually shifts from sensory regions toward emotional and limbic structures.

Figure 4

Longitudinal changes in brain activity underlying spontaneous pain when patients transition from an acute to a chronic back pain state (used with permission from Hashmi et al. [64]). At visits 1 and 2, recovering and persistent subacute back pain (SBP) groups show activation within acute pain regions including the bilateral insula, thalamus, and anterior cingulate cortex. At visits 3 and 4, patients recovering from SBP show no significant activity, whereas increased activation in the medial prefrontal cortex and amygdala is observed in patients with persistent SBP at visit 4.

Limbic and Cortical Involvement

The interaction of limbic–cortical circuits identified by neuroimaging studies is consistent with the concept of close interaction between pain, memory, and learning. In this model, chronic pain is pain that fails to extinguish its memory trace, and instead reflects a state of continuous learning in which the interaction between the prefrontal cortex and limbic learning circuitry is central to the transition to chronicity (Figure 5 ) [130].

Figure 5

A model of the brain circuitry involved in the transition from acute to chronic pain (used with permission from Mansour et al. [130]). ACC, anterior cingulate cortex; Amyg = amygdala; Hippo = hippocampus; lPFC = lateral prefrontal cortex; mPFC = medial prefrontal cortex; NAc = nucleus accumbens.

Transient nociceptive signals mainly evoke acute pain perception through activation of the anterior cingulate cortex and insular cortex. Mansour et al. [130] proposed a model in which limbic–cortical plasticity underlies the shift to chronic pain. In this model, normally any learned associations mediated by the limbic circuitry would be gradually extinguished or unlearned with time [130]. However, the limbic circuitry could be preferentially activated (including the amygdala, hippocampus, and NAc) if the nociceptive signal was persistent and/or intense. These structures are integral to learning and memory. In turn, these pathways shift cortical activity from a predominately nociceptive state to a more emotional one by interacting with the prefrontal cortical circuitry, with pain transitioning to a more emotional state [130]. The limbic circuitry also provides modulatory signals to the cortex, causing functional and anatomical alterations. According to this hypothesis, the persistence of pain-related perceptions is driven by implicit (nondeclarative) and explicit (declarative) memories interacting with subconscious signals. Thus, chronic pain is either “unlearned” or maintained depending on the reaction of the mesolimbic emotional learning circuitry.

Interactions also occur at the spinal level. That is, the spinal cord also exhibits plasticity, and nociceptive inputs that are altered by peripheral and spinal cord sensitization processes impinge on limbic circuitry. Conversely, the altered learning processes in the limbic–cortical circuitry impact descending modulatory pathways, thereby influencing spinal cord responses to nociceptive inputs [130]. Thus, the chronification of pain involves interactions at multiple levels and offers several possible therapeutic targets. Increased understanding of the neurophysiology of chronic pain has provided insight into potential prophylactic and treatment strategies that prevent the establishment of chronic pain pathways or reverse the reorganization that accompanies chronic pain including regions involved in memory and learning.

Prevention and Treatment of Chronic Pain

Ideally, chronic pain should be prevented early by pharmacological and/or psychological interventions to prevent the establishment of the pain-induced plasticity in the nervous system analogous to memory mechanisms. Acute pain, when poorly treated, may lead to chronic pain syndromes; therefore, preemptive analgesia represents a possible approach to preventing chronic pain by blocking peripheral and central nociception [131]. Some evidence favors pharmacologic agents that interfere with molecules involved in CNS plasticity (eg, perioperative nerve block) as effective preemptive analgesics [132–136]; however, controversy remains regarding this approach, and the body of literature focused on preemptive analgesia is limited.

Regarding management of established chronic pain, several clinical neuroimaging studies have shown that effective interventions can restore, at least partially, normal brain structure and function [137, 138]. The reversal of chronic pain may be possible with pharmacologic intervention that prevents or reverses the reorganization that accompanies chronic pain, and a very wide range of targets has been and continues to be evaluated. These targets include specific ion channels, neurotransmitters, and receptors associated with pain signaling pathways [136–141]. In addition, trophic factors that may contribute to changes in the properties of individual neurons and the sprouting of their axons are under investigation [69].

Pharmacologic agents more likely to be encountered in the primary care setting include gabapentin and pregabalin, which bind selectively to α2δ subunits of presynaptic voltage-gated calcium channels and act to inhibit intense neuronal activity by decreasing the calcium-mediated release of glutamate and other neurotransmitters [142–146]. The α2δ calcium channel subunit is thought to mediate the hyperexcitability underlying neuropathic pain [54, 66]. These findings are of interest given the established role of voltage-gated calcium channels in synaptic plasticity in the spinal cord [147] and amygdala [51]. Systematic reviews provide reasonably good evidence for the efficacy of gabapentin and/or pregabalin in chronic pain conditions, including diabetic neuropathy, post-herpetic neuralgia, central neuropathic pain, and fibromyalgia [148, 149]. In addition, mirogabalin, a novel preferentially selective α2δ ligand in development, was well tolerated and demonstrated efficacy for diabetic peripheral neuropathic pain [150, 151].

NMDA receptor antagonists, such as dextromethorphan and ketamine, may have some preemptive analgesic benefit [152]. Ketamine has seen increasing use as an analgesic for acute pain [152], and the literature supports its effectiveness in several forms of chronic pain [153]. In addition to its analgesic properties, likely due to NMDA receptor antagonism, ketamine has rapid-acting antidepressant effects, possibly mediated through an increase in AMPA receptor signaling [97]. Low-dose ketamine infusion has successfully treated refractory neuropathic pain syndromes, which may involve significant depressive comorbidities, most notably complex regional pain syndrome [154, 155]. Interestingly, chronic administration of ketamine appears to lead to memory impairments due to reduced hippocampal activation [156].

Currently, two classes of antidepressants, serotonin and norepinephrine reuptake inhibitors (SNRIs) and tricyclic antidepressants (TCAs), are recommended for the treatment of a variety of chronic pain conditions [157, 158]. SNRIs and TCAs block monoamine uptake, thereby increasing extracellular levels of monoamine neurotransmitters [35]. For SNRIs, activation of the descending modulatory pathway from the RVM and PAG to the dorsal horn neurons and altered norepinephrine signaling and synaptic transmission in the brain are all possible mechanisms [23, 97, 159]. Of note, the antidepressant effect of SNRIs may arise from their effect on synaptic plasticity in the cortex, hippocampus, and striatum, all regions implicated in memory and learning. SNRIs have been successful in treating a range of chronic pain conditions, including diabetic neuropathy, fibromyalgia, and chronic back pain [160–162]. The pharmacologic mechanisms for SNRIs and TCAs are similar [97], and over the last two decades, TCAs have been used to treat a number of chronic pain conditions, most notably neuropathic pain conditions [163, 164].

Psychological approaches that focus on factors that exacerbate or maintain chronic pain, such as maladaptive learned brain responses and failure to extinguish (unlearn) pain responses, are possible with operant behavioral therapy, cognitive behavioral therapy (CBT), and motivational interviewing [165, 166]. Several functional neuroimaging studies have demonstrated that CBT can alter the brain network connectivity that occurs with repeated pain experience [167, 168]. In one randomized fMRI study of patients with chronic pain, functional connectivity between the anterior Default Mode Network and the amygdala/periaqueductal gray region decreased following CBT compared with controls not receiving CBT [168]. Another investigation indicated that CBT in patients with chronic pain resulted in increased gray matter volume in areas that also play a role in learning and memory, including the prefrontal and somatosensory brain regions and hippocampus [169]. Furthermore, increased dorsolateral prefrontal volume was associated with reduced pain catastrophizing [169]. These results add to mounting evidence that CBT can be a valuable treatment option for chronic pain. In addition, techniques such as imagery, mirror training, or the use of virtual reality may help address the maladaptive brain plasticity that accompanies chronic pain [170–173]. Integrative approaches to chronic pain management that consider the patient’s biopsychosocial history include physical activity and exercise, nutrition and supplementation, and yoga [174].

Nonpharmacologic interventions for pain can also play a role in memory. For example, vagal nerve stimulation relieves pain in patients with migraine [175] and some patients with fibromyalgia [176], and can also enhance recognition memory [177]. Acupuncture improves osteoarthritis pain and improves working memory [178, 179].

Exercise (eg, resistance or flexibility exercise, yoga, and tai chi) relieves pain in some patients [180] and also improves memory [181]. The effect of exercise, in particular, on chronic pain is an active area of research. Exercise is thought to exert an analgesic effect through the activation of endogenous systems, particularly the opioidergic, nitrergic, serotoninergic, noradrenergic, and endocannabinoidergic systems, as well as through the release of anti-inflammatory cytokines [182–184]. Recent studies have unraveled the potential of exercise to not only relieve but prevent chronic pain via immune system modulation [185, 186]. Six weeks of voluntary wheel running prevented the full development of allodynia due to chronic constriction injury in rats and caused an increase in levels of the anti-inflammatory cytokine interleukin (IL)-10 compared with sedentary controls [186]. In mice, eight weeks of voluntary wheel running caused an increase in the proportion of macrophages that secrete anti-inflammatory cytokines compared with inflammatory macrophages in the gastrocnemius muscle. Physically active mice were also resistant to acid injection–induced hyperalgesia, and this analgesic effect could be prevented by blockade of IL-10 [185].

Clinical studies show promise for exercise in alleviating chronic pain due to osteoarthritis of the hip and knee [187–189], fibromyalgia [190], intermittent claudication [191], pain of the low back [192–195], neck [196, 197], patella, and femur [198], and pain associated with type 2 diabetes [199]. However, there is some conflicting evidence suggesting that physical activity has no significant effect on pain of the low back [200] or pain due to fibromyalgia [201]. Overall, increasing the sample size of clinical studies could allow stronger conclusions to be drawn about the benefit of exercise for chronic pain patients [180]. It will also be important to study which type of exercise has a positive effect for the specific type of pain condition.

Overall, nonpharmacological interventions can be easily integrated as part of a multidisciplinary, multimodal approach that combines pharmacological, physical, and psychological components tailored to each patient’s needs. Specific noninvasive brain stimulation techniques that induce electrical stimulation to directly alter brain activity, such as repetitive transcranial magnetic stimulation, may have some short-term benefit in chronic pain, although additional evidence is required [170]. More studies are needed to investigate the underlying physiological changes in learning, memory, or pain-related areas of the CNS that accompany the analgesic effects of nonpharmacologic approaches.

Conclusions

In sum, chronic pain can result from a persistent pain stimulus due to injury or disease, but it can also persist after the original injury is healed. Nonetheless, chronic pain is accompanied by the persistence of pain plasticity mechanisms analogous to memory and/or the failure to terminate pain plasticity induced by the original inciting injury. This conceptual framework is consistent with neuroimaging evidence of structural and functional neuroplasticity associated with chronic pain, as well as the large body of clinical and experimental evidence demonstrating anatomical and physiologic overlap with learning and memory. In this context, chronic pain can be conceptualized as the consequence of plastic changes in limbic–cortical circuitry, leading to new learning and reinforced maladaptive plasticity analogous to memory mechanisms that cannot be extinguished due to emotional associations with painful stimuli. Further studies are needed to fully understand the key factors influencing the shift from acute to chronic pain and the changes in learning, memory, or pain-related areas of the CNS that accompany analgesic effects of nonpharmacologic approaches. A better understanding of these mechanisms will aid in the development of novel therapeutic approaches that reverse the pain plasticity and relieve chronic pain.

Pharmacological and psychological approaches that rationally target the processes and mechanisms underlying chronic pain are currently available to clinicians. Additional targets and future approaches to treatment might include novel agents that interfere with receptors and molecules involved in LTP in both subcortical and cortical targets (eg, NMDA receptor, voltage-gated calcium channels and their components, BDNF), as well as nonpharmacological approaches that focus on intensive intervention to prevent consolidation of acute into chronic pain and extinguishing memory-like plasticity associated with chronic pain.

Authors’ Contributions

The authors wrote and revised this manuscript and approved the final draft.

Acknowledgments

Editorial support was provided by Claire Daniele, PhD, and was funded by Daiichi Sankyo, Inc.

References

1

St Sauver
JL
,
Warner
DO
,
Yawn
BP
et al. .
Why patients visit their doctors: Assessing the most prevalent conditions in a defined American population
.
Mayo Clin Proc
2013
;
88
:
56
67
.

2

Treede
RD
,
Rief
W
,
Barke
A
et al. .
A classification of chronic pain for ICD-11
.
Pain
2015
;
156
:
1003
7
.

3

Moriarty
O
,
McGuire
BE
,
Finn
DP
.
The effect of pain on cognitive function: A review of clinical and preclinical research
.
Prog Neurobiol
2011
;
93
3
:
385
404
.

4

Andrew
R
,
Derry
S
,
Taylor
RS
,
Straube
S
,
Phillips
CJ
.
The costs and consequences of adequately managed chronic non-cancer pain and chronic neuropathic pain
.
Pain Pract
2014
;
14
1
:
79
94
.

5

Committee on Advancing Pain Research, Care, and Education; Board on Health Sciences Policy; Institute of Medicine
.
Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research
.
Washington, DC
:
National Academies Press;
2011
.

6

Peppin
JF
,
Schatman
ME
.
Terminology of chronic pain: The need to “level the playing field
.”
J Pain Res
2016
;
9
:
23
4
.

7

Melzack
R
.
From the gate to the neuromatrix
.
Pain
1999
;
82
(
Suppl
1
):
S121
6
.

8

Zhuo
M
.
Cortical excitation and chronic pain
.
Trends Neurosci
2008
;
31
4
:
199
207
.

9

Bourne
S
,
Machado
AG
,
Nagel
SJ
.
Basic anatomy and physiology of pain pathways
.
Neurosurg Clin N Am
2014
;
25
4
:
629
38
.

10

Dubin
AE
,
Patapoutian
A
.
Nociceptors: The sensors of the pain pathway
.
J Clin Invest
2010
;
120
11
:
3760
72
.

11

Reddi
D
,
Curran
N
,
Stephens
R
.
An introduction to pain pathways and mechanisms
.
Br J Hosp Med (Lond)
2013
;
74
(
Suppl
12
):
C188
91
.

12

Swenson
RS
.
Chapter 7: Somatosensory systems. In: Review of Clinical and Functional Neuroscience
.
Hanover, NH
:
Dartmouth Medical School
;
2006
. https://www.dartmouth.edu/~rswenson/NeuroSci/chapter_7.html (accessed January 9, 2019).

13

Cata
JP
,
Weng
HR
,
Chen
JH
,
Dougherty
PM
.
Altered discharges of spinal wide dynamic range neurons and down-regulation of glutamate transporter expression in rats with paclitaxel-induced hyperalgesia
.
Neuroscience
2006
;
138
1
:
329
38
.

14

Maixner
W
,
Dubner
R
,
Bushnell
MC
,
Kenshalo
DR
Jr
,
Oliveras
JL
.
Wide-dynamic-range dorsal horn neurons participate in the encoding process by which monkeys perceive the intensity of noxious heat stimuli
.
Brain Res
1986
;
374
2
:
385
8
.

15

Roberts
WJ
,
Foglesong
ME
.
Spinal recordings suggest that wide-dynamic-range neurons mediate sympathetically maintained pain
.
Pain
1988
;
34
3
:
289
304
.

16

Price
DD
.
Psychological and neural mechanisms of the affective dimension of pain
.
Science
2000
;
288
5472
:
1769
72
.

17

Apkarian
AV
,
Bushnell
MC
,
Treede
RD
,
Zubieta
JK
.
Human brain mechanisms of pain perception and regulation in health and disease
.
Eur J Pain
2005
;
9
4
:
463
84
.

18

Ossipov
MH
,
Dussor
GO
,
Porreca
F
.
Central modulation of pain
.
J Clin Invest
2010
;
120
11
:
3779
87
.

19

Ossipov
MH
,
Morimura
K
,
Porreca
F
.
Descending pain modulation and chronification of pain
.
Curr Opin Support Palliat Care
2014
;
8
2
:
143
51
.

20

Schweinhardt
P
,
Bushnell
MC
.
Neuroimaging of pain: Insights into normal and pathological pain mechanisms
.
Neurosci Lett
2012
;
520
2
:
129
30
.

21

Tracey
I
,
Johns
E
.
The pain matrix: Reloaded or reborn as we image tonic pain using arterial spin labelling
.
Pain
2010
;
148
3
:
359
60
.

22

Morgan
MM
,
Whittier
KL
,
Hegarty
DM
,
Aicher
SA
.
Periaqueductal gray neurons project to spinally projecting GABAergic neurons in the rostral ventromedial medulla
.
Pain
2008
;
140
2
:
376
86
.

23

Pertovaara
A
.
Noradrenergic pain modulation
.
Prog Neurobiol
2006
;
80
2
:
53
83
.

24

Taves
S
,
Berta
T
,
Chen
G
,
Ji
RR
.
Microglia and spinal cord synaptic plasticity in persistent pain
.
Neural Plast
2013
;
2013
:
753656
.

25

Tiwari
V
,
Guan
Y
,
Raja
SN
.
Modulating the delicate glial-neuronal interactions in neuropathic pain: Promises and potential caveats
.
Neurosci Biobehav Rev
2014
;
45
:
19
27
.

26

Ji
RR
,
Berta
T
,
Nedergaard
M
.
Glia and pain: Is chronic pain a gliopathy?
Pain
2013
;
154
(
Suppl 1
):
S10
28
.

27

Liou
JT
,
Lee
CM
,
Day
YJ
.
The immune aspect in neuropathic pain: Role of chemokines
.
Acta Anaesthesiol Taiwan
2013
;
51
3
:
127
32
.

28

Scholz
J
,
Woolf
CJ
.
The neuropathic pain triad: Neurons, immune cells and glia
.
Nat Neurosci
2007
;
10
11
:
1361
8
.

29

Watkins
LR
,
Hutchinson
MR
,
Ledeboer
A
et al. .
Glia as the “bad guys”: Implications for improving clinical pain control and the clinical utility of opioids
.
Brain Behav Immun
2007
;
21
2
:
131
46
.

30

Bradesi
S
.
Role of spinal cord glia in the central processing of peripheral pain perception
.
Neurogastroenterol Motil
2010
;
22
5
:
499
511
.

31

Gosselin
RD
,
Suter
MR
,
Ji
RR
,
Decosterd
I
.
Glial cells and chronic pain
.
Neuroscientist
2010
;
16
5
:
519
31
.

32

Kuner
R
.
Central mechanisms of pathological pain
.
Nat Med
2010
;
16
11
:
1258
66
.

33

Latremoliere
A
,
Woolf
CJ
.
Central sensitization: A generator of pain hypersensitivity by central neural plasticity
.
J Pain
2009
;
10
9
:
895
926
.

34

Woolf
CJ
.
Evidence for a central component of post-injury pain hypersensitivity
.
Nature
1983
;
306
5944
:
686
8
.

35

Yaksh
TL
,
Woller
SA
,
Ramachandran
R
,
Sorkin
LS
.
The search for novel analgesics: Targets and mechanisms
.
F1000Prime Reports
2015
;
7
56
:
1
27
.

36

Woolf
CJ
,
Thompson
SW
.
The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states
.
Pain
1991
;
44
3
:
293
9
.

37

Dickenson
AH
,
Sullivan
AF
.
Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C fibre stimulation
.
Neuropharmacology
1987
;
26
8
:
1235
8
.

38

Murakami
M
,
Nakagawasai
O
,
Suzuki
T
et al. .
Antinociceptive effect of different types of calcium channel inhibitors and the distribution of various calcium channel alpha 1 subunits in the dorsal horn of spinal cord in mice
.
Brain Res
2004
;
1024
(
1–2
):
122
9
.

39

Ferguson
AR
,
Huie
JR
,
Crown
ED
et al. .
Maladaptive spinal plasticity opposes spinal learning and recovery in spinal cord injury
.
Front Physiol
2012
;
3
:
399
.

40

Woolf
CJ
.
Central sensitization: Implications for the diagnosis and treatment of pain
.
Pain
2011
;
152
(
suppl
3
):
S2
15
.

41

Sivilotti
LG
,
Thompson
SW
,
Woolf
CJ
.
Rate of rise of the cumulative depolarization evoked by repetitive stimulation of small-caliber afferents is a predictor of action potential windup in rat spinal neurons in vitro
.
J Neurophysiol
1993
;
69
5
:
1621
31
.

42

Nadel
L
,
Hardt
O
.
Update on memory systems and processes
.
Neuropsychopharmacology
2011
;
36
1
:
251
73
.

43

Orsini
CA
,
Maren
S
.
Neural and cellular mechanisms of fear and extinction memory formation
.
Neurosci Biobehav Rev
2012
;
36
7
:
1773
802
.

44

Byrne
JH
.
Chapter 7: Learning and memory. In: Neuroscience Online: An Electronic Textbook for the Neurosciences
.
The University of Texas Health Science Center at Houston (UTHealth)
;
1997
. https://nba.uth.tmc.edu/neuroscience/m/s4/chapter07.html (accessed January 9, 2019).

45

Jin
J
,
Maren
S
.
Prefrontal-hippocampal interactions in memory and emotion
.
Front Syst Neurosci
2015
;
9
:
170
.

46

Richter-Levin
G
.
The amygdala, the hippocampus, and emotional modulation of memory
.
Neuroscientist
2004
;
10
1
:
31
9
.

47

Richter-Levin
G
,
Akirav
I
.
Amygdala-hippocampus dynamic interaction in relation to memory
.
Mol Neurobiol
2000
;
22
(
1–3
):
11
20
.

48

Blair
HT
,
Schafe
GE
,
Bauer
EP
,
Rodrigues
SM
,
LeDoux
JE
.
Synaptic plasticity in the lateral amygdala: A cellular hypothesis of fear conditioning
.
Learn Mem
2001
;
8
5
:
229
42
.

49

Magerl
W
,
Hansen
N
,
Treede
RD
,
Klein
T
.
The human pain system exhibits higher-order plasticity (metaplasticity)
.
Neurobiol Learn Mem
2018
;
154
112
20
.

50

Lamprecht
R
,
LeDoux
J
.
Structural plasticity and memory
.
Nat Rev Neurosci
2004
;
5
1
:
45
54
.

51

Bauer
EP
,
Schafe
GE
,
LeDoux
JE
.
NMDA receptors and L-type voltage-gated calcium channels contribute to long-term potentiation and different components of fear memory formation in the lateral amygdala
.
J Neurosci
2002
;
22
12
:
5239
49
.

52

Rison
RA
,
Stanton
PK
.
Long-term potentiation and N-methyl-D-aspartate receptors: Foundations of memory and neurologic disease?
Neurosci Biobehav Rev
1995
;
19(4)
:
533
52
.

53

Nelson
TJ
,
Alkon
DL
.
Molecular regulation of synaptogenesis during associative learning and memory
.
Brain Res
2015
;
1621
:
239
51
.

54

Li
KW
,
Yu
YP
,
Zhou
C
et al. .
Calcium channel alpha2delta1 proteins mediate trigeminal neuropathic pain states associated with aberrant excitatory synaptogenesis
.
J Biol Chem
2014
;
289
10
:
7025
37
.

55

Sterpenich
V
,
Albouy
G
,
Darsaud
A
et al. .
Sleep promotes the neural reorganization of remote emotional memory
.
J Neurosci
2009
;
29
16
:
5143
52
.

56

Bird
GC
,
Lash
LL
,
Han
JS
et al. .
Protein kinase A-dependent enhanced NMDA receptor function in pain-related synaptic plasticity in rat amygdala neurones
.
J Physiol
2005
;
564
(
Pt 3
):
907
21
.

57

Fu
Y
,
Han
J
,
Ishola
T
et al. .
PKA and ERK, but not PKC, in the amygdala contribute to pain-related synaptic plasticity and behavior
.
Mol Pain
2008
;
4
:
26
.

58

Goncalves
L
,
Silva
R
,
Pinto-Ribeiro
F
et al. .
Neuropathic pain is associated with depressive behaviour and induces neuroplasticity in the amygdala of the rat
.
Exp Neurol
2008
;
213
1
:
48
56
.

59

Mutso
AA
,
Radzicki
D
,
Baliki
MN
et al. .
Abnormalities in hippocampal functioning with persistent pain
.
J Neurosci
2012
;
32
17
:
5747
56
.

60

Neugebauer
V
,
Li
W
,
Bird
GC
et al. .
Synaptic plasticity in the amygdala in a model of arthritic pain: Differential roles of metabotropic glutamate receptors 1 and 5
.
J Neurosci
2003
;
23
1
:
52
63
.

61

Ploghaus
A
,
Narain
C
,
Beckmann
CF
et al. .
Exacerbation of pain by anxiety is associated with activity in a hippocampal network
.
J Neurosci
2001
;
21
24
:
9896
903
.

62

Kayser
V
,
Elfassi
IE
,
Aubel
B
et al. .
Mechanical, thermal and formalin-induced nociception is differentially altered in 5-HT1A-/-, 5-HT1B-/-, 5-HT2A-/-, 5-HT3A-/- and 5-HTT-/- knock-out male mice
.
Pain
2007
;
130
3
:
235
48
.

63

Seyedabadi
M
,
Fakhfouri
G
,
Ramezani
V
,
Mehr
SE
,
Rahimian
R
.
The role of serotonin in memory: Interactions with neurotransmitters and downstream signaling
.
Exp Brain Res
2014
;
232
3
:
723
38
.

64

Hashmi
JA
,
Baliki
MN
,
Huang
L
et al. .
Shape shifting pain: Chronification of back pain shifts brain representation from nociceptive to emotional circuits
.
Brain
2013
;
136
9
:
2751
68
.

65

Baliki
MN
,
Petre
B
,
Torbey
S
et al. .
Corticostriatal functional connectivity predicts transition to chronic back pain
.
Nat Neurosci
2012
;
15
8
:
1117
9
.

66

Li
CY
,
Zhang
XL
,
Matthews
EA
et al. .
Calcium channel alpha2delta1 subunit mediates spinal hyperexcitability in pain modulation
.
Pain
2006
;
125
(
1–2
):
20
34
.

67

Chen
JL
,
Margolis
DJ
,
Stankov
A
et al. .
Pathway-specific reorganization of projection neurons in somatosensory cortex during learning
.
Nat Neurosci
2015
;
18
8
:
1101
8
.

68

Schafe
GE
,
Nader
K
,
Blair
HT
,
LeDoux
JE
.
Memory consolidation of Pavlovian fear conditioning: A cellular and molecular perspective
.
Trends Neurosci
2001
;
24
9
:
540
6
.

69

Costigan
M
,
Scholz
J
,
Woolf
CJ
.
Neuropathic pain: A maladaptive response of the nervous system to damage
.
Annu Rev Neurosci
2009
;
32
:
1
32
.

70

Nabavi
S
,
Fox
R
,
Proulx
CD
et al. .
Engineering a memory with LTD and LTP
.
Nature
2014
;
511
7509
:
348
52
.

71

Kato
A
,
Punnakkal
P
,
Pernia-Andrade
AJ
et al. .
Endocannabinoid-dependent plasticity at spinal nociceptor synapses
.
J Physiol
2012
;
590
19
:
4717
33
.

72

Kleim
JA
,
Hogg
TM
,
VandenBerg
PM
et al. .
Cortical synaptogenesis and motor map reorganization occur during late, but not early, phase of motor skill learning
.
J Neurosci
2004
;
24
3
:
628
33
.

73

Rosenberg
T
,
Gal-Ben-Ari
S
,
Dieterich
DC
et al. .
The roles of protein expression in synaptic plasticity and memory consolidation
.
Front Mol Neurosci
2014
;
7
:
86
.

74

Kandel
ER
.
The molecular biology of memory storage: A dialog between genes and synapses
.
Biosci Rep
2004
;
24
(
4–5
):
475
522
.

75

Jog
MS
,
Kubota
Y
,
Connolly
CI
,
Hillegaart
V
,
Graybiel
AM
.
Building neural representations of habits
.
Science
1999
;
286
5445
:
1745
9
.

76

Knowlton
BJ
,
Mangels
JA
,
Squire
LR
.
A neostriatal habit learning system in humans
.
Science
1996
;
273
5280
:
1399
402
.

77

Lessard
N
,
McNally-Gagnon
A
,
Dubuc
MJ
et al. .
Preserved priming effect in individuals with schizophrenia: Cues towards rehabilitation
.
Cogn Neuropsychiatry
2015
;
20
6
:
512
25
.

78

Lee
KH
,
Mathews
PJ
,
Reeves
AM
et al. .
Circuit mechanisms underlying motor memory formation in the cerebellum
.
Neuron
2015
;
86
2
:
529
40
.

79

Gordon
GR
,
Baimoukhametova
DV
,
Hewitt
SA
et al. .
Norepinephrine triggers release of glial ATP to increase postsynaptic efficacy
.
Nat Neurosci
2005
;
8
8
:
1078
86
.

80

Panatier
A
,
Theodosis
DT
,
Mothet
JP
et al. .
Glia-derived D-serine controls NMDA receptor activity and synaptic memory
.
Cell
2006
;
125
4
:
775
84
.

81

Perea
G
,
Araque
A
.
Properties of synaptically evoked astrocyte calcium signal reveal synaptic information processing by astrocytes
.
J Neurosci
2005
;
25
9
:
2192
203
.

82

Czopka
T
,
Ffrench-Constant
C
,
Lyons
DA
.
Individual oligodendrocytes have only a few hours in which to generate new myelin sheaths in vivo
.
Dev Cell
2013
;
25
6
:
599
609
.

83

Dutta
R
,
Chang
A
,
Doud
MK
et al. .
Demyelination causes synaptic alterations in hippocampi from multiple sclerosis patients
.
Ann Neurol
2011
;
69
3
:
445
54
.

84

Schmued
LC
,
Raymick
J
,
Paule
MG
,
Dumas
M
,
Sarkar
S
.
Characterization of myelin pathology in the hippocampal complex of a transgenic mouse model of Alzheimer’s disease
.
Curr Alzheimer Res
2013
;
10
:
30
7
.

85

Blom
SM
,
Pfister
JP
,
Santello
M
,
Senn
W
,
Nevian
T
.
Nerve injury-induced neuropathic pain causes disinhibition of the anterior cingulate cortex
.
J Neurosci
2014
;
34
17
:
5754
64
.

86

Yang
JX
,
Hua
L
,
Li
YQ
et al. .
Caveolin-1 in the anterior cingulate cortex modulates chronic neuropathic pain via regulation of NMDA receptor 2B subunit
.
J Neurosci
2015
;
35
1
:
36
52
.

87

Goffer
Y
,
Xu
D
,
Eberle
SE
et al. .
Calcium-permeable AMPA receptors in the nucleus accumbens regulate depression-like behaviors in the chronic neuropathic pain state
.
J Neurosci
2013
;
33
48
:
19034
44
.

88

Ziv
M
,
Tomer
R
,
Defrin
R
,
Hendler
T
.
Individual sensitivity to pain expectancy is related to differential activation of the hippocampus and amygdala
.
Hum Brain Mapp
2010
;
31
:
326
38
.

89

Aarse
J
,
Herlitze
S
,
Manahan-Vaughan
D
.
The requirement of BDNF for hippocampal synaptic plasticity is experience-dependent
.
Hippocampus
2016
;
26
6
:
739
51
.

90

Guo
W
,
Robbins
MT
,
Wei
F
et al. .
Supraspinal brain-derived neurotrophic factor signaling: A novel mechanism for descending pain facilitation
.
J Neurosci
2006
;
26
1
:
126
37
.

91

Barlow
RL
,
Alsio
J
,
Jupp
B
et al. .
Markers of serotonergic function in the orbitofrontal cortex and dorsal raphe nucleus predict individual variation in spatial-discrimination serial reversal learning
.
Neuropsychopharmacology
2015
;
40
7
:
1619
30
.

92

Buhot
MC
,
Wolff
M
,
Benhassine
N
et al. .
Spatial learning in the 5-HT1B receptor knockout mouse: Selective facilitation/impairment depending on the cognitive demand
.
Learn Mem
2003
;
10
:
466
77
.

93

Buhot
MC
,
Wolff
M
,
Savova
M
et al. .
Protective effect of 5-HT1B receptor gene deletion on the age-related decline in spatial learning abilities in mice
.
Behav Brain Res
2003
;
142
(
1–2
):
135
42
.

94

Leiser
SC
,
Li
Y
,
Pehrson
AL
et al. .
Serotonergic regulation of prefrontal cortical circuitries involved in cognitive processing: A review of individual 5-HT receptor mechanisms and concerted effects of 5-HT receptors exemplified by the multimodal antidepressant vortioxetine
.
ACS Chem Neurosci
2015
;
6
7
:
970
86
.

95

Puig
MV
,
Gulledge
AT
.
Serotonin and prefrontal cortex function: Neurons, networks, and circuits
.
Mol Neurobiol
2011
;
44
3
:
449
64
.

96

Rodriguez
JJ
,
Noristani
HN
,
Verkhratsky
A
.
The serotonergic system in ageing and Alzheimer’s disease
.
Prog Neurobiol
2012
;
99
1
:
15
41
.

97

Doan
L
,
Manders
T
,
Wang
J
.
Neuroplasticity underlying the comorbidity of pain and depression
.
Neural Plast
2015
;
2015
:
504691
.

98

Millan
MJ
.
Descending control of pain
.
Prog Neurobiol
2002
;
66
6
:
355
474
.

99

Apkarian
AV
,
Sosa
Y
,
Sonty
S
et al. .
Chronic back pain is associated with decreased prefrontal and thalamic gray matter density
.
J Neurosci
2004
;
24
46
:
10410
5
.

100

Good
CD
,
Johnsrude
IS
,
Ashburner
J
et al. .
A voxel-based morphometric study of ageing in 465 normal adult human brains
.
Neuroimage
2001
;
14
1
:
21
36
.

101

Resnick
SM
,
Pham
DL
,
Kraut
MA
,
Zonderman
AB
,
Davatzikos
C
.
Longitudinal magnetic resonance imaging studies of older adults: A shrinking brain
.
J Neurosci
2003
;
23
8
:
3295
301
.

102

Blankstein
U
,
Chen
J
,
Diamant
NE
,
Davis
KD
.
Altered brain structure in irritable bowel syndrome: Potential contributions of pre-existing and disease-driven factors
.
Gastroenterology
2010
;
138
5
:
1783
9
.

103

Geha
PY
,
Baliki
MN
,
Harden
RN
et al. .
The brain in chronic CRPS pain: Abnormal gray-white matter interactions in emotional and autonomic regions
.
Neuron
2008
;
60
4
:
570
81
.

104

Kim
JH
,
Suh
SI
,
Seol
HY
et al. .
Regional grey matter changes in patients with migraine: A voxel-based morphometry study
.
Cephalalgia
2008
;
28
6
:
598
604
.

105

Kuchinad
A
,
Schweinhardt
P
,
Seminowicz
DA
et al. .
Accelerated brain gray matter loss in fibromyalgia patients: Premature aging of the brain?
J Neurosci
2007
;
27(15)
:
4004
7
.

106

Luerding
R
,
Weigand
T
,
Bogdahn
U
,
Schmidt-Wilcke
T
.
Working memory performance is correlated with local brain morphology in the medial frontal and anterior cingulate cortex in fibromyalgia patients: Structural correlates of pain-cognition interaction
.
Brain
2008
;
131
(
Pt 12
):
3222
31
.

107

Rodriguez-Raecke
R
,
Niemeier
A
,
Ihle
K
,
Ruether
W
,
May
A
.
Brain gray matter decrease in chronic pain is the consequence and not the cause of pain
.
J Neurosci
2009
;
29
44
:
13746
50
.

108

Schmidt-Wilcke
T
,
Gänßbauer
S
,
Neuner
T
,
Bogdahn
U
,
May
A
.
Subtle grey matter changes between migraine patients and healthy controls
.
Cephalalgia
2008
;
28
1
:
1
4
.

109

Schmidt-Wilcke
T
,
Leinisch
E
,
Gänssbauer
S
et al. .
Affective components and intensity of pain correlate with structural differences in gray matter in chronic back pain patients
.
Pain
2006
;
125
(
1–2
):
89
97
.

110

Schmidt-Wilcke
T
,
Leinisch
E
,
Straube
A
et al. .
Gray matter decrease in patients with chronic tension type headache
.
Neurology
2005
;
65
9
:
1483
6
.

111

Schmidt-Wilcke
T
,
Luerding
R
,
Weigand
T
et al. .
Striatal grey matter increase in patients suffering from fibromyalgia—A voxel-based morphometry study
.
Pain
2007
;
132
(
Suppl
1
):
S109
16
.

112

Schweinhardt
P
,
Kuchinad
A
,
Pukall
CF
,
Bushnell
MC
.
Increased gray matter density in young women with chronic vulvar pain
.
Pain
2008
;
140
3
:
411
9
.

113

Baliki
MN
,
Chialvo
DR
,
Geha
PY
et al. .
Chronic pain and the emotional brain: Specific brain activity associated with spontaneous fluctuations of intensity of chronic back pain
.
J Neurosci
2006
;
26
47
:
12165
73
.

114

Baliki
MN
,
Geha
PY
,
Fields
HL
,
Apkarian
AV
.
Predicting value of pain and analgesia: Nucleus accumbens response to noxious stimuli changes in the presence of chronic pain
.
Neuron
2010
;
66
1
:
149
60
.

115

Farmer
MA
,
Chanda
ML
,
Parks
EL
et al. .
Brain functional and anatomical changes in chronic prostatitis/chronic pelvic pain syndrome
.
J Urol
2011
;
186
1
:
117
24
.

116

Geha
PY
,
Baliki
MN
,
Chialvo
DR
et al. .
Brain activity for spontaneous pain of postherpetic neuralgia and its modulation by lidocaine patch therapy
.
Pain
2007
;
128
(
1–2
):
88
100
.

117

Geha
PY
,
Baliki
MN
,
Wang
X
et al. .
Brain dynamics for perception of tactile allodynia (touch-induced pain) in postherpetic neuralgia
.
Pain
2008
;
138
3
:
641
56
.

118

Gussew
A
,
Rzanny
R
,
Gullmar
D
,
Scholle
HC
,
Reichenbach
JR
.
1H-MR spectroscopic detection of metabolic changes in pain processing brain regions in the presence of non-specific chronic low back pain
.
Neuroimage
2011
;
54
2
:
1315
23
.

119

Gustin
SM
,
Peck
CC
,
Wilcox
SL
et al. .
Different pain, different brain: Thalamic anatomy in neuropathic and non-neuropathic chronic pain syndromes
.
J Neurosci
2011
;
31
16
:
5956
64
.

120

Hashmi
JA
,
Baliki
MN
,
Huang
L
et al. .
Lidocaine patch (5%) is no more potent than placebo in treating chronic back pain when tested in a randomised double blind placebo controlled brain imaging study
.
Mol Pain
2012
;
8
:
29
.

121

Valdes
M
,
Collado
A
,
Bargallo
N
et al. .
Increased glutamate/glutamine compounds in the brains of patients with fibromyalgia: A magnetic resonance spectroscopy study
.
Arthritis Rheum
2010
;
62
6
:
1829
36
.

122

Urenjak
J
,
Williams
SR
,
Gadian
DG
,
Noble
M
.
Proton nuclear magnetic resonance spectroscopy unambiguously identifies different neural cell types
.
J Neurosci
1993
;
13
3
:
981
9
.

123

Jutzeler
CR
,
Curt
A
,
Kramer
JL
.
Relationship between chronic pain and brain reorganization after deafferentation: A systematic review of functional MRI findings
.
Neuroimage Clin
2015
;
9
:
599
606
.

124

Nardone
R
,
Holler
Y
,
Brigo
F
et al. .
Functional brain reorganization after spinal cord injury: Systematic review of animal and human studies
.
Brain Res
2013
;
1504
:
58
73
.

125

Cauda
F
,
Sacco
K
,
Duca
S
et al. .
Altered resting state in diabetic neuropathic pain
.
PLoS One
2009
;
4
2
:
e4542
.

126

Malinen
S
,
Vartiainen
N
,
Hlushchuk
Y
et al. .
Aberrant temporal and spatial brain activity during rest in patients with chronic pain
.
Proc Natl Acad Sci U S A
2010
;
107
14
:
6493
7
.

127

Napadow
V
,
LaCount
L
,
Park
K
et al. .
Intrinsic brain connectivity in fibromyalgia is associated with chronic pain intensity
.
Arthritis Rheum
2010
;
62
8
:
2545
55
.

128

Tagliazucchi
E
,
Balenzuela
P
,
Fraiman
D
,
Chialvo
DR
.
Brain resting state is disrupted in chronic back pain patients
.
Neurosci Lett
2010
;
485
1
:
26
31
.

129

Baliki
MN
,
Baria
AT
,
Apkarian
AV
.
The cortical rhythms of chronic back pain
.
J Neurosci
2011
;
31
39
:
13981
90
.

130

Mansour
AR
,
Farmer
MA
,
Baliki
MN
,
Apkarian
AV
.
Chronic pain: The role of learning and brain plasticity
.
Restor Neurol Neurosci
2014
;
32
1
:
129
39
.

131

Grape
S
,
Tramer
MR
.
Do we need preemptive analgesia for the treatment of postoperative pain?
Best Pract Res Clin Anaesthesiol
2007
;
21(1)
:
51
63
.

132

Bailey
M
,
Corcoran
T
,
Schug
S
,
Toner
A
.
Perioperative lidocaine infusions for the prevention of chronic postsurgical pain: A systematic review and meta-analysis of efficacy and safety
.
Pain
2018
;
159
9
:
1696
704
.

133

Al-Mujadi
H
,
A-Refai
AR
,
Katzarov
MG
et al. .
Preemptive gabapentin reduces postoperative pain and opioid demand following thyroid surgery
.
Can J Anaesth
2006
;
53
3
:
268
73
.

134

Buvanendran
A
,
Kroin
JS
,
Della Valle
CJ
et al. .
Perioperative oral pregabalin reduces chronic pain after total knee arthroplasty: A prospective, randomized, controlled trial
.
Anesth Analg
2010
;
110
1
:
199
207
.

135

Lee
JK
,
Chung
KS
,
Choi
CH
.
The effect of a single dose of preemptive pregabalin administered with COX-2 inhibitor: A trial in total knee arthroplasty
.
J Arthroplasty
2015
;
30
1
:
38
42
.

136

Tuchman
M
,
Barrett
JA
,
Donevan
S
,
Hedberg
TG
,
Taylor
CP
.
Central sensitization and Ca(V)alpha(2)delta ligands in chronic pain syndromes: Pathologic processes and pharmacologic effect
.
J Pain
2010
;
11
12
:
1241
9
.

137

Ceko
M
,
Shir
Y
,
Ouellet
JA
et al. .
Partial recovery of abnormal insula and dorsolateral prefrontal connectivity to cognitive networks in chronic low back pain after treatment
.
Hum Brain Mapp
2015
;
36
:
2075
92
.

138

Seminowicz
DA
,
Wideman
TH
,
Naso
L
et al. .
Effective treatment of chronic low back pain in humans reverses abnormal brain anatomy and function
.
J Neurosci
2011
;
31
20
:
7540
50
.

139

Borsook
D
,
Hargreaves
R
,
Bountra
C
,
Porreca
F
.
Lost but making progress–where will new analgesic drugs come from?
Sci Transl Med
2014
;
6(249)
249sr3
.

140

Sałat
K
,
Kowalczyk
P
,
Gryzło
B
,
Jakubowska
A
,
Kulig
K
.
New investigational drugs for the treatment of neuropathic pain
.
Expert Opin Investig Drugs
2014
;
23
8
:
1093
104
.

141

Schaible
HG
.
Emerging concepts of pain therapy based on neuronal mechanisms
.
Handb Exp Pharmacol
2015
;
227
:
1
14
.

142

Kumar
N
,
Laferriere
A
,
Yu
JS
,
Leavitt
A
,
Coderre
TJ
.
Evidence that pregabalin reduces neuropathic pain by inhibiting the spinal release of glutamate
.
J Neurochem
2010
;
113
2
:
552
61
.

143

Bockbrader
HN
,
Wesche
D
,
Miller
R
et al. .
A comparison of the pharmacokinetics and pharmacodynamics of pregabalin and gabapentin
.
Clin Pharmacokinet
2010
;
49
10
:
661
9
.

144

Field
MJ
,
Cox
PJ
,
Stott
E
et al. .
Identification of the alpha2-delta-1 subunit of voltage-dependent calcium channels as a molecular target for pain mediating the analgesic actions of pregabalin
.
Proc Natl Acad Sci U S A
2006
;
103
46
:
17537
42
.

145

Gee
NS
,
Brown
JP
,
Dissanayake
VU
et al. .
The novel anticonvulsant drug, gabapentin (Neurontin), binds to the alpha2delta subunit of a calcium channel
.
J Biol Chem
1996
;
271
10
:
5768
76
.

146

Harris
RE
,
Napadow
V
,
Huggins
JP
et al. .
Pregabalin rectifies aberrant brain chemistry, connectivity, and functional response in chronic pain patients
.
Anesthesiology
2013
;
119
6
:
1453
64
.

147

Liu
XG
,
Zhou
LJ
.
Long-term potentiation at spinal C-fiber synapses: A target for pathological pain
.
Curr Pharm Des
2015
;
21
7
:
895
905
.

148

Moore
RA
,
Wiffen
PJ
,
Derry
S
,
Toelle
T
,
Rice
AS
.
Gabapentin for chronic neuropathic pain and fibromyalgia in adults
.
Cochrane Database Syst Rev
2014
;
4
:
CD007938
.

149

Wiffen
PJ
,
Derry
S
,
Moore
RA
et al. .
Antiepileptic drugs for neuropathic pain and fibromyalgia—an overview of Cochrane reviews
.
Cochrane Database Syst Rev
2013
;
11
:
CD010567
.

150

Merante
D
,
Rosenstock
J
,
Sharma
U
et al. .
Efficacy of mirogabalin (DS-5565) on patient-reported pain and sleep interference in patients with diabetic neuropathic pain: Secondary outcomes of a phase II proof-of-concept study
.
Pain Med
2017
;
18
11
:
2198
207
.

151

Vinik
A
,
Rosenstock
J
,
Sharma
U
et al. .
Efficacy and safety of mirogabalin (DS-5565) for the treatment of diabetic peripheral neuropathic pain: A randomized, double-blind, placebo- and active comparator-controlled, adaptive proof-of-concept phase 2 study
.
Diabetes Care
2014
;
37
12
:
3253
61
.

152

Peltoniemi
MA
,
Hagelberg
NM
,
Olkkola
KT
,
Saari
TI
.
Ketamine: A review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy
.
Clin Pharmacokinet
2016
;
55
9
:
1059
77
.

153

Tawfic
QA
.
A review of the use of ketamine in pain management
.
J Opioid Manag
2013
;
9
5
:
379
88
.

154

Correll
GE
,
Maleki
J
,
Gracely
EJ
,
Muir
JJ
,
Harbut
RE
.
Subanesthetic ketamine infusion therapy: A retrospective analysis of a novel therapeutic approach to complex regional pain syndrome
.
Pain Med
2004
;
5
3
:
263
75
.

155

Sigtermans
MJ
,
van Hilten
JJ
,
Bauer
MC
et al. .
Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1
.
Pain
2009
;
145
3
:
304
11
.

156

Morgan
CJ
,
Dodds
CM
,
Furby
H
et al. .
Long-term heavy ketamine use is associated with spatial memory impairment and altered hippocampal activation
.
Front Psychiatry
2014
;
5
:
149
.

157

CYMBALTA
.
CYMBALTA (Duloxetine Delayed-Release Capsules) for Oral Use [package insert]. Indianapolis
, IN:
Lilly USA
,
LLC
;
2016
.

158

Finnerup
NB
,
Attal
N
,
Haroutounian
S
et al. .
Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis
.
Lancet Neurol
2015
;
14
2
:
162
73
.

159

Bernik
M
,
Sampaio
TP
,
Gandarela
L
.
Fibromyalgia comorbid with anxiety disorders and depression: Combined medical and psychological treatment
.
Curr Pain Headache Rep
2013
;
17
:
358
.

160

Arnold
LM
.
Duloxetine and other antidepressants in the treatment of patients with fibromyalgia
.
Pain Med
2007
;
8
(
Suppl
2
):
S63
74
.

161

Ormseth
MJ
,
Scholz
BA
,
Boomershine
CS
.
Duloxetine in the management of diabetic peripheral neuropathic pain
.
Patient Prefer Adherence
2011
;
5
:
343
56
.

162

Schukro
RP
,
Oehmke
MJ
,
Geroldinger
A
et al. .
Efficacy of duloxetine in chronic low back pain with a neuropathic component: A randomized, double-blind, placebo-controlled crossover trial
.
Anesthesiology
2016
;
124
1
:
150
8
.

163

Max
MB
,
Lynch
SA
,
Muir
J
et al. .
Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy
.
N Engl J Med
1992
;
326
19
:
1250
6
.

164

Moore
RA
,
Derry
S
,
Aldington
D
,
Cole
P
,
Wiffen
PJ
.
Amitriptyline for neuropathic pain and fibromyalgia in adults
.
Cochrane Database Syst Rev
2012
;
12
CD008242
.

165

Molton
IR
,
Graham
C
,
Stoelb
BL
,
Jensen
MP
.
Current psychological approaches to the management of chronic pain
.
Curr Opin Anaesthesiol
2007
;
20
5
:
485
9
.

166

Osborne
TL
,
Raichle
KA
,
Jensen
MP
.
Psychologic interventions for chronic pain
.
Phys Med Rehabil Clin N Am
2006
;
17
2
:
415
33
.

167

Kucyi
A
,
Salomons
TV
,
Davis
KD
.
Cognitive behavioral training reverses the effect of pain exposure on brain network activity
.
Pain
2016
;
157
9
:
1895
904
.

168

Shpaner
M
,
Kelly
C
,
Lieberman
G
et al. .
Unlearning chronic pain: A randomized controlled trial to investigate changes in intrinsic brain connectivity following cognitive behavioral therapy
.
Neuroimage Clin
2014
;
5
:
365
76
.

169

Seminowicz
DA
,
Shpaner
M
,
Keaser
ML
et al. .
Cognitive-behavioral therapy increases prefrontal cortex gray matter in patients with chronic pain
.
J Pain
2013
;
14
12
:
1573
84
.

170

Ramachandran
VS
,
Altschuler
EL
.
The use of visual feedback, in particular mirror visual feedback, in restoring brain function
.
Brain
2009
;
132
7
:
1693
710
.

171

Bowering
KJ
,
O’Connell
NE
,
Tabor
A
et al. .
The effects of graded motor imagery and its components on chronic pain: A systematic review and meta-analysis
.
J Pain
2013
;
14
1
:
3
13
.

172

Cacchio
A
,
De Blasis
E
,
Necozione
S
,
di Orio
F
,
Santilli
V
.
Mirror therapy for chronic complex regional pain syndrome type 1 and stroke
.
N Engl J Med
2009
;
361
6
:
634
6
.

173

Malloy
KM
,
Milling
LS
.
The effectiveness of virtual reality distraction for pain reduction: A systematic review
.
Clin Psychol Rev
2010
;
30
8
:
1011
8
.

174

Bonakdar
RA
.
Integrative pain management
.
Med Clin North Am
2017
;
101
5
:
987
1004
.

175

Chakravarthy
K
,
Chaudhry
H
,
Williams
K
,
Christo
PJ
.
Review of the uses of vagal nerve stimulation in chronic pain management
.
Curr Pain Headache Rep
2015
;
19
12
:
54
.

176

Lange
G
,
Janal
MN
,
Maniker
A
et al. .
Safety and efficacy of vagus nerve stimulation in fibromyalgia: A phase I/II proof of concept trial
.
Pain Med
2011
;
12
9
:
1406
13
.

177

Clark
KB
,
Naritoku
DK
,
Smith
DC
,
Browning
RA
,
Jensen
RA
.
Enhanced recognition memory following vagus nerve stimulation in human subjects
.
Nat Neurosci
1999
;
2
1
:
94
8
.

178

Bussell
J
.
The effect of acupuncture on working memory and anxiety
.
J Acupunct Meridian Stud
2013
;
6
5
:
241
6
.

179

Egorova
N
,
Gollub
RL
,
Kong
J
.
Repeated verum but not placebo acupuncture normalizes connectivity in brain regions dysregulated in chronic pain
.
Neuroimage Clin
2015
;
9
:
430
5
.

180

Geneen
LJ
,
Moore
RA
,
Clarke
C
et al. .
Physical activity and exercise for chronic pain in adults: An overview of Cochrane reviews
.
Cochrane Database Syst Rev
2017
;
1
:
CD011279
.

181

Roig
M
,
Nordbrandt
S
,
Geertsen
SS
,
Nielsen
JB
.
The effects of cardiovascular exercise on human memory: A review with meta-analysis
.
Neurosci Biobehav Rev
2013
;
37
8
:
1645
66
.

182

Da Silva Santos
R
,
Galdino
G
.
Endogenous systems involved in exercise-induced analgesia
.
J Physiol Pharmacol
2018
;
69
1
:
3
13
.

183

Bodnar
RJ
,
Kelly
DD
,
Spiaggia
A
,
Ehrenberg
C
,
Glusman
M
.
Dose-dependent reductions by naloxone of analgesia induced by cold-water stress
.
Pharmacol Biochem Behav
1978
;
8
6
:
667
72
.

184

Colt
EW
,
Wardlaw
SL
,
Frantz
AG
.
The effect of running on plasma beta-endorphin
.
Life Sci
1981
;
28
14
:
1637
40
.

185

Leung
A
,
Gregory
NS
,
Allen
LA
,
Sluka
KA
.
Regular physical activity prevents chronic pain by altering resident muscle macrophage phenotype and increasing interleukin-10 in mice
.
Pain
2016
;
157
1
:
70
9
.

186

Grace
PM
,
Fabisiak
TJ
,
Green-Fulgham
SM
et al. .
Prior voluntary wheel running attenuates neuropathic pain
.
Pain
2016
;
157
9
:
2012
23
.

187

Fransen
M
,
McConnell
S
,
Hernandez-Molina
G
,
Reichenbach
S
.
Exercise for osteoarthritis of the hip
.
Cochrane Database Syst Rev
2014
;
4
:
CD007912
.

188

Fransen
M
,
McConnell
S
,
Harmer
AR
et al. .
Exercise for osteoarthritis of the knee
.
Cochrane Database Syst Rev
2015
;
1
:
CD004376
.

189

Bartels
EM
,
Juhl
CB
,
Christensen
R
et al. .
Aquatic exercise for the treatment of knee and hip osteoarthritis
.
Cochrane Database Syst Rev
2016
;
3
CD005523
.

190

Busch
AJ
,
Webber
SC
,
Richards
RS
et al. .
Resistance exercise training for fibromyalgia
.
Cochrane Database Syst Rev
2013
;
12
:
CD010884
.

191

Lane
R
,
Harwood
A
,
Watson
L
,
Leng
GC
.
Exercise for intermittent claudication
.
Cochrane Database Syst Rev
2017
;
12
:
CD000990
.

192

Hayden
JA
,
van Tulder
MW
,
Malmivaara
A
,
Koes
BW
.
Exercise therapy for treatment of non-specific low back pain
.
Cochrane Database Syst Rev
2005
;
3
CD000335
.

193

Yamato
TP
,
Maher
CG
,
Saragiotto
BT
et al. .
Pilates for low back pain
.
Cochrane Database Syst Rev
2015
;
7
:
CD010265
.

194

Cortell-Tormo
JM
,
Sanchez
PT
,
Chulvi-Medrano
I
et al. .
Effects of functional resistance training on fitness and quality of life in females with chronic nonspecific low-back pain
.
J Back Musculoskelet Rehabil
2018
;
31
1
:
95
105
.

195

Bodes Pardo
G
,
Lluch Girbes
E
,
Roussel
NA
et al. .
Pain neurophysiology education and therapeutic exercise for patients with chronic low back pain: A single-blind randomized controlled trial
.
Arch Phys Med Rehabil
2018
;
99
2
:
338
47
.

196

Galindez-Ibarbengoetxea
X
,
Setuain
I
,
Ramirez-Velez
R
et al. .
Short-term effects of manipulative treatment versus a therapeutic home exercise protocol for chronic cervical pain: A randomized clinical trial
.
J Back Musculoskelet Rehabil
2018
;
31
1
:
133
45
.

197

Uluğ
N
,
Yılmaz
ÖT
,
Kara
M
,
Özçakar
L
.
Effects of pilates and yoga in patients with chronic neck pain: A sonographic study
.
J Rehabil Med
2018
;
50
1
:
80
5
.

198

van der Heijden
RA
,
Lankhorst
NE
,
van Linschoten
R
,
Bierma-Zeinstra
SM
,
van Middelkoop
M
.
Exercise for treating patellofemoral pain syndrome
.
Cochrane Database Syst Rev
2015
;
1
:
CD010387
.

199

Schmid
AA
,
Atler
KE
,
Malcolm
MP
et al. .
Yoga improves quality of life and fall risk-factors in a sample of people with chronic pain and type 2 diabetes
.
Complement Ther Clin Pract
2018
;
31
369
73
.

200

McCaskey
MA
,
Wirth
B
,
Schuster-Amft
C
,
de Bruin
ED
.
Postural sensorimotor training versus sham exercise in physiotherapy of patients with chronic non-specific low back pain: An exploratory randomised controlled trial
.
PLoS One
2018
;
13(3)
e0193358
.

201

Bidonde
J
,
Busch
AJ
,
Webber
SC
et al. .
Aquatic exercise training for fibromyalgia
.
Cochrane Database Syst Rev
2014
;
10
:
CD011336
.

Author notes

Funding sources: The authors received no direct financial support for the writing, review, or publishing of this article. Editorial support was funded by Daiichi Sankyo, Inc.

Conflicts of interest: John Peppin served as a consultant for OneSourceRegulatory, YourEncore, and Janssen Pharmaceuticals and served as a consultant and speaker for Ferring Pharmaceuticals. Bill McCarberg served as an advisor for Pfizer, Collegium, Daiichi Sankyo, and Pernix and served on the speaker’s bureau for Collegium. He has stock holdings in Johnson and Johnson, Biospecifics Technologies, Galena, and Collegium.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)