IMR Press / JIN / Volume 23 / Issue 5 / DOI: 10.31083/j.jin2305089
Open Access Review
The Complexity of Neuropathic Pain and Central Sensitization: Exploring Mechanisms and Therapeutic Prospects
Show Less
1 Orthopedics Department, Lanzhou University Second Hospital, 730000 Lanzhou, Gansu, China
2 Orthopaedics Key Laboratory of Gansu Province, 730000 Lanzhou, Gansu, China
3 Stomatology Department, Lanzhou University Second Hospital, 730000 Lanzhou, Gansu, China
4 The Second Clinical Medical College, Lanzhou University, 730000 Lanzhou, Gansu, China
*Correspondence: NWei2023@163.com (Wei Nan)
J. Integr. Neurosci. 2024, 23(5), 89; https://doi.org/10.31083/j.jin2305089 (registering DOI)
Submitted: 3 October 2023 | Revised: 23 November 2023 | Accepted: 12 December 2023 | Published: 25 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Neuropathic pain is a common pain syndrome, which seriously affects the quality of life of patients. The mechanism of neuropathic pain is complex. Peripheral tissue injury can trigger peripheral sensitization; however, what really plays a key role is the sensitization of the central nervous system. Central sensitization is a key factor in the perception of chronic pain. Central sensitization refers to the increased sensitivity of the central nervous system to pain treatment, which is related to the change of the functional connection mode of the neural network. The current study aims to reveal the basic molecular mechanisms of central sensitization, including the involvement of P2 purine X4 receptor and brain-derived neurotrophic factor. In terms of treatment, although there are drugs and physical therapy, the accuracy of targeting is limited and the efficacy needs to be further improved. Future therapeutic strategies may involve the development of new drugs designed to specifically inhibit the central sensitization process. This article focuses on the effector molecules involved in central sensitization, aiming to elucidate the pathogenesis of neuropathic pain and provide a basis for the development of more effective treatment models.

Keywords
neuropathic pain
central sensitization
P2 purine X4 receptor
brain-derived neurotrophic factor
1. Introduction

Neuropathic pain (NP) refers to a series of diseases of the peripheral nervous system caused by injury or disease of the nervous system part that usually transmits sensory information, which seriously affects the quality of life of patients [1, 2]. A variety of diseases, such as trigeminal neuralgia, postherpetic neuralgia, diabetic neuropathy-related pain, post-stroke central pain, etc., can cause NP [1, 3]. With the aging of the global population, NP has gradually evolved into a more urgent clinical problem [4]. At present, the clinical treatment of NP encounters many obstacles: opioids provide limited analgesic effects and cause many adverse reactions [5]; non-steroidal anti-inflammatory drugs (NSAIDs) and antidepressants are facing a prominent problem of drug resistance [6]; biological agents such as monoclonal antibodies targeting recombinant nerve growth factor (NGF) have shown the advantages of targeted therapy, but concerns about their safety still exist [7]. Therefore, it is imperative to develop new, safe and effective NP therapeutic drugs.

In the pathogenesis of NP, although peripheral tissue damage can induce peripheral sensitization and enhance pain signals, more and more studies have emphasized that central sensitization is the basic mechanism for NP maintenance [8, 9]. Central sensitization refers to the increasing responsiveness of the central nervous system to pain stimuli, which is a key determinant of maintaining various pain states [10]. Under the condition of central sensitization, pain-related central regions exhibit a series of pathological changes across cellular and molecular levels, including enhanced neuronal excitability, synaptic reorganization and weakened efficacy of intrinsic pain suppression mechanisms, which eventually lead to enhanced pain response and tolerance [11, 12, 13]. The comprehensive exploration of the mechanism of central sensitization and the formulation of treatment methods for central sensitization may open up new avenues for the treatment of NP [14]. In addition, central sensitization is also an important therapeutic target in chronic, painful, inflammatory and non-inflammatory rheumatic diseases and central spondylitis [15, 16]. However, the understanding of the molecular basis of central sensitization is still limited, and its intricate cellular and molecular effect networks need to be further elucidated.

Central sensitization plays a key role in the pathogenesis of NP [17]. In recent years, the research focus of neuropathic pain has gradually shifted to the key effector molecules of central sensitization, and great progress has been made. Nevertheless, a comprehensive understanding of the precise molecular mechanism of central sensitization is still limited to ongoing research and needs to be further elucidated [18]. This article elaborates the specific mechanism of central sensitization in NP, including changes at the molecular level, changes at the cellular level, and regulation of neural networks. At the same time, the animal model of NP is summarized to promote the research in the field of NP.

2. Clinical Manifestations of Neuropathic Pain

NP can be manifested as spontaneous pain, hyperalgesia and malformed pain [19]. Persistent spontaneous pain seriously affects the quality of life of patients [20]. Hyperalgesia refers to peripheral and central sensitization [21]. The pain caused by deformity means the injury of pain pathway [22]. These signs suggest the presence of pain amplification and increased central sensitivity.

2.1 Spontaneous Pains

Spontaneous pain is the main manifestation of neuropathic pain, which is a pain sensation without obvious causal relationship. It often persists and exhibits a severe nature, seriously affecting the patient’s sleep pattern and overall quality of life [23]. According to the location of the lesion, it can be divided into central spontaneous pain and peripheral spontaneous pain [24, 25]. Central spontaneous pain often occurs after stroke or brain injury, manifested as unprovoked pain in the facial area or affecting half of the limbs [26]. Peripheral spontaneous pain is common, including trigeminal neuralgia, facial pain, postherpetic neuralgia, and limb pain caused by diabetic peripheral neuropathy [27, 28]. These pain symptoms can last for months or even years, significantly affecting the patient’s sleep patterns, emotional state and overall quality of life [29]. Drug therapy for spontaneous pain is still insufficient, and a combination of antidepressants, antiepileptics, and opioids is often required to relieve symptoms [30].

2.2 Hyperalgesia and Allodynia

Hyperalgesia refers to the amplification response to harmful stimuli, which can increase the body’s pain response to harmful stimuli [31]. This pain often occurs in the initial stage of NP, caused by peripheral and central sensitization [32]. Hyperalgesia is particularly common after surgery or in the initial stage of nerve injury, which may last for weeks to months [33]. Allodynia refers to the reaction of ordinary or mild stimulation, such as gentle touch/pressure can cause pain response [34]. Hyperalgesia can be managed with corticosteroids, non-steroidal anti-inflammatory drugs, and nerve blockers [35]. Identifying and effectively managing hyperalgesia helps to avoid the development of central sensitization and prevent pain from progressing to a chronic state.

2.3 Phantom Limb Pain

Phantom limb pain refers to the phenomenon that patients still feel pain or other feelings related to the limb when they lose the limb [36]. This phenomenon usually occurs in patients undergoing surgery or accidental limb amputation. Even if the limb has been amputated or lost, the patient still feels the presence of the limb, and sometimes even feels discomfort such as pain, itching, and touch [37]. Phantom Limb Pain may be related to the plasticity of the central nervous system [38]. After losing a limb, the brain may be able to adapt to the change by reorganizing neural circuits [39]. This reorganization may lead to confusion of neural signals, resulting in a sense of phantom limb pain, that is, central sensitization.

3. The Mechanism of Neuropathic Pain

While peripheral tissue damage can induce peripheral sensitization, it is central sensitization that assumes greater significance in augmenting and sustaining pain perception [40]. Central sensitization correlates with heightened neuronal excitability, synaptic reorganization, and diminished inhibitory control [41]. Studies have shown that the signaling pathways involved in P2 purine X4 receptor (P2X4R) and brain-derived neurotrophic factor tyrosine kinase receptor B (BDNF-Trk B) are the basic components of central sensitization [42].

3.1 Neuropathic Pain Caused by Peripheral Nerve Injury
3.1.1 Pathological Changes after Peripheral Nerve Injury

Peripheral nerve injury can significantly trigger the release of inflammatory mediators such as tumor necrosis factor α, substance P and interleukin, activate and sensitize the peripheral pain afferent pathway, and is the main catalyst for the pathogenesis of NP [43]. These inflammatory mediators can directly affect the peripheral nerve conduction pathway. In addition, they can also stimulate the aggregation and activation of a variety of immune cells at the site of tissue damage, further exacerbating the inflammatory response [44, 45]. Peripheral inflammation can induce chemoreceptor sensitization, reduce the activation threshold of peripheral nerves, increase the production and release of stimulating neurotransmitters, promote abnormal discharge, and ultimately lead to atypical amplification and sensitization of peripheral afferent pathways [46]. In the peripheral inflammatory pain model, the inflammatory response induced by immune cell infiltration can directly trigger and sensitize the terminals of sensory neurons [47]. In addition, inflammatory mediators such as ATP, substance P and leukotrienes released from the injured site can bind to the corresponding receptors at the end of sensory neurons and enhance the responsiveness of neurons to stimuli [48]. In addition, these inflammatory mediators can stimulate the pathological discharge of peripheral nerves and maintain a long-term excitatory state [49]. Studies have shown that in an inflammatory environment, the expression of peripheral nerve voltage-gated Na+ channels such as sodium ion channels (Na V1.8) is increased, and the frequency of nerve impulses is increased, resulting in spontaneous discharges and hyperalgesia [50].

3.1.2 Peripheral Nerve Injury Leads to Sensitization of Pain Pathway

Abnormal discharges from damaged nerves into the spinal cord can increase the excitability of sensory neurons in the spinal dorsal horn [51]. This encompasses heightened activity of N-methyl-D-aspartic acid receptors (NMDAR), elevated α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) phosphorylation, and reduced synthesis and release of inhibitory neurotransmitters like glycine (Gly) and γ-aminobutyric acid (GABA) [52]. These changes lead to increased excitability of the spinal dorsal horn neural network, pathological sensitization of peripheral afferent pathways, and enhanced transmission of peripheral pain input signals to the central nervous system [53]. In the peripheral injury model, high-frequency afferent stimulation can induce persistent enhancement of spinal dorsal horn neurons, which means a direct central sensitization process [54]. The electrophysiological recording of spinal dorsal horn neurons also showed an increase in baseline discharge frequency, reflecting the increase in neuronal excitability [55]. A meta-analysis conducted by Zhao et al. [56] showed that the release of substance P and calcitonin gene-related peptide (CGRP) can promote central sensitization and lead to the development of headache. In addition, microglial cells are activated, enhancing the signal regulation of neurons [57, 58]. Therefore, over-activated peripheral inputs can affect the spinal cord network in a variety of ways, eventually leading to enhanced central processing and pain amplification (Fig. 1).

Fig. 1.

Peripheral sensitization mechanism map of neuropathic pain. Following peripheral nerve or tissue injury, immune cells (like mast cells and basophils) liberate a range of inflammatory mediators, encompassing interleukin-6 (IL-6), interleukin-1β (IL-1β), substance P, tumor necrosis factor-α (TNF-α), insulin-like growth factor (IGF), and nerve growth factor (NGF). These inflammatory mediators orchestrate peripheral sensitization via the subsequent mechanisms: (1) Activation of TRPV1: Inflammatory factors stimulate TRPV1 channels in peripheral nerves, inducing an augmentation of calcium (Ca2+) influx and elevated synthesis and release of NGF; (2) Activation of Nav1.8: Inflammatory factors trigger Nav1.8 channel activity in peripheral nerves, resulting in the influx of sodium ions (Na+). The internal Na+ flow additionally activates the voltage-gated calcium channel, intensifying Ca2+ flow; (3) NGF-induced signaling pathway: NGF stimulates TrkA receptors on peripheral nerves, initiating PKC and PI3K pathways. TrkA receptor activation amplifies Nav1.8 activity, subsequently fostering sensitization of peripheral receptors; furthermore, inflammatory factors discharged by immune cells can activate their Toll-like receptors (TLR), consequently setting off the MAPK pathway and the NF-κB pathway, which further elevates the secretion of inflammatory factors. These cumulative effects result in heightened activity of peripheral afferent pathways, rendering them more receptive to pain stimuli. This subsequently conveys an increased volume of pain information to the central nervous system, thereby establishing a fundamental groundwork for the establishment of central sensitization. PGE2, Prostaglandin E2; COX-2, Cyclooxygenase-2; TRPV1, Transient Receptor Potential Cation Channel Subfamily V Member 1; PKC, Protein Kinase C; PI3K, Phosphoinositide 3-Kinase; TrkA, Tropomyosin Receptor Kinase A; NF-κB, Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells; MAPK, Mitogen-Activated Protein Kinase.

3.1.3 The Mechanism of Pain Amplification after Peripheral Nerve Injury

The enhancement of excitability of spinal dorsal horn neurons will amplify the incoming pain stimulation information, which will eventually lead to the establishment of pain amplification effect [59]. Increased excitability of spinal dorsal horn neurons is associated with NMDAR activation, spinal neural network remodeling, and decreased levels of inhibitory neurotransmitters [60]. At the spinal level, we observed that Amyloid-β (A β) light tactile afferent fibers activate tactile response neurons and trigger pain responses, indicating abnormal activation and amplification effects [61]. NMDAR antagonists can significantly improve pain amplification, suggesting that NMDAR is involved in this process [62]. There are a series of neurotransmitters involved in regulation in the spinal cord, including inhibitory neurotransmitters such as Gly and GABA [63]. These inhibitory neurotransmitters can suppress excessive spinal reflex excitation. If the synthesis or release of these inhibitory neurotransmitters in the spinal cord is insufficient, the inhibitory control of the spinal cord network will be weakened, thereby promoting the excessive excitement of the spinal reflex pathway and triggering a certain pathological response. Persistent stimulation of pain-related receptors can lead to activation and positive regulation of synaptic NMDAR in the spinal dorsal horn, resulting in amplification of pain signals transmitted to the brain (central sensitization) [64]. Pain amplification leads to a strong output signal to a slight stimulus, which transmits a strong pain sensation to the brain and induces a strong pain response in the individual. Moreover, abnormal stimulation of spinal cord neurons can also react to peripheral tissues, forming a positive feedback loop of pain.

3.2 The Central Mechanism of Neuropathic Pain
3.2.1 The Concept and Characteristics of Central Sensitization

Central sensitization is the key mechanism to maintain neuropathic pain. The overactive state of spinal microglia and astrocytes is one of the key factors to activate central sensitization [65, 66]. Central sensitization mainly occurs in the pain processing center of the spinal dorsal horn and brainstem, which may last for months to years. The intensity of central sensitization is related to the amplitude and duration of pain [67]. In addition, functional magnetic resonance imaging (MRI) studies have shown that patients with chronic pain have increased and extensive activation of central regions during acupuncture pain stimulation, which means that the central nervous system is more sensitive in pain information processing [68].

3.2.2 Molecular Mechanism of Central Sensitization

Central sensitization includes cellular and molecular mechanisms, including increasing neuronal excitability, altering synaptic plasticity, and attenuating inhibitory neuromodulation [69, 70]. Activated microglia can release a large amount of neuroactive substances and pro-inflammatory cytokines [71]. Decreased release of inhibitory neurotransmitters such as GABA and glycine can also impair the inhibitory control of neural networks [72]. In addition, activated protein kinases such as extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) induce changes in downstream gene expression and cell activity [73, 74]. These factors increase the excitability of the central nervous network, thereby more effectively amplifying and transmitting pain signals. At the molecular level, during central sensitization, the concentration of excitatory amino acid neurotransmitters, such as Glutamate (Glu), increased, while the level of inhibitory neurotransmitters, such as GABA, decreased [75, 76]. The generation and maintenance of central sensitization is also related to the following mechanisms: (1) Increased release of inflammatory cytokines directly regulates the excitability of neurons. Central sensitization can increase the release of pro-inflammatory factors, which can directly act on neurons and regulate their excitability [77, 78]; (2) Activate glial cells and enhance the signal regulation of neurons [79]. In the state of central sensitization, microglia and astrocytes are activated to release cytokines and chemokines, enhance the signal regulation of neurons, and further promote central sensitization [80, 81]; (3) Calcium signaling pathway is activated, affecting neuronal excitability [82]; (4) Increased synaptic connection strength, cell structure remodeling, and changes in neural network sensitivity [83]; (5) The hypothalamus-adrenal cortex axis is activated, affecting neuroendocrine processes [84, 85]. Changes in functional connectivity between these brain regions also help to shape the effects of central sensitization. Brain imaging studies have shown that functional connectivity between different brain regions is enhanced during central sensitization, which promotes the processing and persistence of pain [86]. Although a large number of studies have conducted in-depth studies on key molecules in central sensitization, such as P2X4R, Brain-Derived Neurotrophic Factor (BDNF) and NMDAR, their precise mechanisms and complex interactions between different pain types still need to be further elucidated through recent animal experiments.

3.3 Maintain Central Sensitization Related Signaling Pathways
3.3.1 P2X4 Receptor Pathway

High expression of P2X4R is an important aspect of central sensitization [87]. The activation of P2X4R increases the excitability of central neurons, triggers downstream signaling cascades, induces the release of inflammatory factors, and produces an escalating hyperalgesia loop [88]. Therefore, P2X4R is a key effector molecule that connects peripheral input and central sensitization. In different animal models of neuropathic pain, the expression of P2X4R in glial cells is increased, and inhibition of P2X4R can significantly reduce hyperalgesia and spontaneous pain [89, 90]. The activation of P2X4R not only directly triggers the increase of intracellular calcium concentration and the increase of central nervous system excitability, but also initiates downstream protein kinases such as ERK and JNK, induces the release of a series of inflammatory factors and cell active substances, and finally leads to the progressive hyperalgesia loop in the central nervous system [91, 92, 93]. Activation of P2X4R in spinal microglia releases BDNF, which can convert Gamma-Aminobutyric Acid Type A Receptor (GABAA) receptor-mediated inhibition into excitation, which is related to the excessive excitability of neurons in the spinal dorsal horn [94, 95, 96]. In addition, the activation of P2X4R promotes the release of ATP and other signaling molecules in glial cells, thus expanding the amplification and regulation of neuronal signals by glial cells [97].

The downstream signaling pathway activated by P2X4R plays a multifaceted role in central sensitization [98]. For example, P2X4R can trigger the activation of Src tyrosine kinase, promote NMDAR phosphorylation and enhance its function [99]. When P2X4R is over-activated, Ca2+ influx increases, prompting protein kinase C (PKC) signaling cascade activation [100]. PKC can phosphorylate transient receptor potential vanilloid 1 (TRPV1), enhance its responsiveness to stimulation, and reduce its activation threshold [101]. In addition, Ca2+ influx caused by P2X4R activation triggers the Mitogen-Activated Protein Kinase (MAPK)/ERK pathway, drives ERK phosphorylation, and then induces the activation of nuclear transcription factor Cyclic Adenosine Monophosphate (cAMP) response element-binding protein (CREB) and the expression of pain-related genes [102]. The synergistic activation of these signaling pathways eventually leads to increased excitability of central neurons and changes in synaptic plasticity. In summary, P2X4R promotes abnormal activation and remodeling of the central nervous network by participating in multiple downstream signaling pathways. It is a key molecular medium connecting peripheral input and central sensitization.

3.3.2 BDNF-TrkB Pathway

BDNF is an important neurotrophic factor, which activates intracellular signaling pathways such as MAPK/ERK and phosphoinositide 3-kinase/protein Kinase B (PI3K/Akt) through its high affinity receptor tropomyosin receptor kinase B (TrkB), and regulates the growth, differentiation and synaptic connection of neurons [103, 104]. Activated glial cells secrete BDNF and lack effective neural regulation mechanisms. Excessive BDNF in the spinal cord may promote the development of maladaptive plasticity by activating the TrkB signaling pathway, promote central sensitization and strengthen the circuit, thus contributing to the maintenance of chronic pain [95, 105, 106]. Activation of BDNF-TrkB signaling induces AMPAR and NMDAR phosphorylation and amplifies Glu receptor-mediated excitatory effects [107, 108]. BDNF first promotes TrkB homodimerize and induces the self-phosphorylation of the binding site, which can activate intracellular signal transduction mediated by BDNF and promote the functional changes in synaptic connection of neurons [109, 110]. In addition, BDNF can activate downstream signaling molecules such as CREB and promote the secretion of pro-inflammatory factors [111].

Activation of the downstream effects of BDNF-TrkB signaling pathway promotes different central sensitization processes:

(1) The binding of BDNF with TrkB receptor can also result in the activation of PKC, which phosphorylates GABAA increasing their presence in the cell surface [112].

(2) BDNF can also promote the phosphorylation of ERK, and ERK can also phosphorylate potassium channels, leading to neuronal depolarization [113, 114].

(3) BDNF promotes the expression of AMPAR, which makes neurotransmitter Glu produce more significant excitatory effect [115].

(4) BDNF can up-regulate the amplification of glial cells to neuronal signals [116].

In conclusion, activation of BDNF-TrkB signaling enhances neuronal excitability, alters synaptic plasticity, and maintains central sensitization (Fig. 2).

Fig. 2.

The molecular mechanism of central sensitization formation and maintenance in neuropathic pain. Primary afferent neurons in the spinal dorsal horn release neurotransmitters including Glu, IL-1β, substance P, ATP, TNF-α, NGF, and others. Among these, Glu and IL-1β activate NMDAR on secondary neurons, causing Ca2+ influx. Simultaneously, NMDAR activates AMPAR, leading to reduced GABA release from inhibitory interneurons. ATP can activate P2X4R on secondary neurons, intensifying NMDAR activation and leading to downstream activation of c-Jun N-terminal kinase (JNK), P38, and MAPK. These mechanisms primarily contribute to the development of central sensitization and synaptic remodeling. Additionally, IL-1β, ATP, and TNF-α released by primary neurons can directly activate microglial P2X4R, leading to the release of BDNF and increased IL-1β levels. Furthermore, BDNF activates TrkB on secondary neurons, triggering MAPK/extracellular signal-regulated kinase (ERK) and PI3K/Akt-cAMP response element binding protein (CREB) signaling pathways, ultimately enhancing NMDAR activation. Moreover, BDNF can activate phosphatidylcholine-specific phospholipase C (PLC-γ) on secondary neurons, subsequently activating downstream diacylglycerol (DAG) and inositol triphosphate (IP3) signaling, exacerbating Ca2+ influx. Additionally, ATP can activate the P2X7 receptor (P2X7R) on astrocytes, inducing the release of further ATP. Glu can stimulate the metabotropic glutamate receptor (mGluR) on astrocytes, leading to increased release of IL-1β and TNF-α. The activation of microglia and astrocytes is crucial for maintaining and exacerbating central sensitization during the process of central sensitization; Glu, Glutamate; MAPK, Mitogen-Activated Protein Kinase; NMDAR, N-methyl-D-aspartic acid receptors; NO, nitric oxide; Gly, glycine; GABA, γ-aminobutyric acid; PI3K, Phosphoinositide 3-Kinase; TrkA, Tropomyosin Receptor Kinase A; AMPAR, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors; BDNF, Brain- Derived Neurotrophic Factor; TrkB, Tropomyosin Receptor Kinase B.

4. Animal Model of Neuropathic Pain

The study of neuropathic pain mostly uses animal models, including peripheral nerve injury, spinal cord injury, central nervous injury, gene knockout model [117, 118]. However, these models have obvious advantages and limitations. In order to enhance their clinical relevance, further optimization of these models is essential.

4.1 Peripheral Nerve Injury Model

In the field of neuropathic pain, peripheral nerve injury models are widely used and have become the main animal models for studying neuropathic pain [119, 120]. Peripheral nerve injury models mainly include:

(1) Chronic constriction injury (CCI) model of sciatic nerve. The CCI model was first proposed by Bennett et al. [121] in 1998. The model included the use of 4-0 chrome catgut to ligate four 1 mm-interval narrowing rings around the sciatic nerve trunk. In the CCI model, significant pain-related behavioral changes were observed within 24 hours after surgery, including mechanical hyperalgesia, thermal hyperalgesia and cold hyperalgesia. These are accompanied by spontaneous pain-like behaviors, including foot contraction, licking the affected limb, and raising the affected limb [122, 123]. However, the main limitation of the CCI model is the challenge of precisely controlling the shrinkage strength. At present, the best contraction intensity is considered to induce a slight tremor in the calf muscles.

(2) Sciatic nerve partial ligation model (PSL). The PSL model was proposed by Seltzer et al. [124] in 1990. The model included the use of a No.8 silk thread and a microbend needle to tightly ligature the proximal 1/3 to 1/2 of the sciatic nerve trunk. Pain behaviors associated with the PSL model can occur within 24 h after surgery and last for 9 weeks [125]. Although this model induces mild injury and usually avoids self-harm or autophagic limbs, controlling the type and number of injured nerve fibers is challenging. The model exhibits huge individual differences, limited repeatability, and significant difficulties in regulating these aspects. It is worth noting that the pain behavior induced by the PSL model tends to disappear within 3–5 weeks and cannot be observed for a long time. In addition, compared with the CCI and sciatic nerve injury (SNI) models, the pain threshold in the PSL model changed less.

(3) Spinal nerve partial ligation model (SNI). The SNI model was developed by Decosterd and Woolf [126] as an extension of the PSL model. Specifically, the SNI model included the use of 6-0 silk sutures to tightly ligature the proximal ends of the tibial and common peroneal nerves. In addition, the distal end of the nerve was sliced and ligated. The distal end of the nerve stump was removed by 2 mm to hinder the reconnection between the stumps and ensure the preservation of the sural nerve. The model has the advantages of good repeatability, consistent nerve fiber type and number of damage, and minimal individual differences [127].

(4) Spinal nerve ligation model (SNL). The SNL model proposed by Ho Kim and Mo Chung [125] includes complete ligation of L5 and L6 spinal nerves with 3-0 silk thread [128]. In this SNL model, the damaged nerve fibers showed homogeneity. It is worth noting that the change of mechanical pain threshold is more obvious than other models. The control of the ligation site and intensity is direct, individual differences are small, and autophagy will not occur [122].

(5) Chronic compressive injury model of spinal nerve root. The dorsal root ganglion chronic compression model developed by Hu and Xing [129] aims to simulate chronic low back pain caused by clinical conditions such as intervertebral foramen stenosis, intervertebral disc herniation, or root ganglion compression by tumors [130]. Other peripheral nerve injury models include L5 spinal nerve root ligation model (L5VRT) and complete sciatic nerve transection model (CST). At present, these two nerve pain models are rarely used.

Peripheral nerve injury models, including CCI, PSL, SNI, SNL and spinal nerve root chronic compression injury models, have wide application value as animal models in the field of neuropathic pain research. It is worth noting that the SNI model stands out because of its robust repeatability and strong damage consistency, and has obvious advantages. This model can effectively promote the research and long-term observation of neuropathic pain.

4.2 Spinal Cord Injury Models

In view of the increased incidence of neuropathic pain after spinal cord injury, it is imperative to understand its potential pathogenesis [131]. This model, commonly known as Allen’s model, includes spinal cord exposure, fixation, and subsequent vertical strikes to induce spinal cord injury [132]. In the spinal cord injury model, it is feasible to prolong the duration of mechanical pain and thermal hyperalgesia [133]. This model provides an opportunity to study the effects of central mechanisms in pain management and central sensitization induced by spinal cord injury. Nevertheless, the creation of spinal cord injury models involves trauma and lacks precision; the resulting severe lower limb paralysis hinders post-impact behavioral assessment. At the same time, autophagy and self-mutilation in the area below the injury site tend to be manifested in the later stages of the disease.

4.3 Central Pain Model after Stroke

About 8% of stroke patients experience post-stroke pain, and the incidence of pain in patients with hemorrhagic stroke is higher [134]. Therefore, the central post-stroke pain model (CPSP), especially the thalamic hemorrhagic stroke model, provides a feasible method for the study of central neuropathic pain. The CPSP model was established by injecting a specific volume of normal saline infused with type IV collagenase into the ventral posterolateral nucleus of the thalamus to induce local bleeding and simulate the clinical hemorrhagic stroke scene. The main features of the CPSP model include mechanical hyperalgesia and cold hyperalgesia, while the thermal pain threshold remains relatively unaffected, possibly due to the degree of tissue damage [135]. The central injury model helps to study the brain’s regulation of pain processing and changes in pain perception due to changes in related neural networks. However, it is more complicated to achieve accurate brain region localization within the model.

Some animal pain models have shown limitations, which makes it necessary to refine models that closely mimic human physiological characteristics and disease progression. This is still an important way of contemporary research. Although the peripheral model provides insight into the peripheral mechanism, it is not sufficient to fully simulate the clinical scenario. The spinal cord injury model allows for the observation of central effects, although the injury is more severe. The post-stroke central pain model allows direct examination of the brain’s involvement in pain, but accurate brain localization has proven to be more complex. In general, these animal models are of great value for analyzing the mechanism of neuropathic pain and designing treatment methods. However, it still needs to be further refined to enhance its applicability in clinical pain research.

5. Clinical Treatment of Neuropathic Pain

In clinical practice, the efficacy of drug therapy for NP is still limited, while biological agents and physical therapy also show inherent limitations [136]. Treatment satisfaction needs to be further improved, because there is still a significant gap in achieving the desired therapeutic effect. Looking forward to the future, new drugs for key molecules of central sensitization should be developed to improve the level of treatment.

5.1 Pharmacological Treatment

At present, drug therapy is the most important means of clinical treatment of NP. Commonly prescribed medications include tricyclic antidepressants (e.g., amitriptyline) and other antidepressants such as duloxetine, antiepileptic drugs (e.g., gabapentin and pregabalin), and opioids [137, 138]. The main mechanism of these drugs involves the regulation of various neurotransmitters and receptors in the central nervous system to produce analgesic effects. Specifically, tricyclic antidepressants can block the reuptake of norepinephrine and 5-hydroxytryptamine, thereby enhancing inhibitory signal transduction between synapses [139]; antiepileptic drugs enhance GABA-mediated inhibitory effects [140, 141]; opioid drugs mainly activate opioid receptors, inhibit calcium channels and adenylate cyclase, thereby reducing central excitability [142, 143]. In addition, some new drugs targeting key aspects of central sensitization have shown good therapeutic potential. For example, NMDAR antagonist ketamine and P2X4R antagonist oxidized ATP (oATP) [144, 145]. The treatment of NP in the elderly usually involves a variety of methods, including drug therapy, physical therapy, psychological support and behavioral therapy [146, 147]. In terms of drug selection, potential adverse event risks need to be considered, especially in view of other health problems and drug interactions that may exist in older persons. Antidepressants such as tricyclic antidepressants (such as amitriptyline) and antiepileptic drugs (such as gabapentin and pregabalin) may lead to decreased cardiac function, cognitive decline, balance problems and sleepiness in the elderly [148, 149]. In addition, cardiovascular adverse events associated with antidepressants also need to be considered. Anesthetic drugs (such as morphine or hydromorphine) can also be used to relieve neuropathic pain in the elderly, but the use of opioids can lead to cognitive decline, constipation, respiratory depression and other problems, and there is a risk of abuse and addiction [150, 151].

These emerging drugs have the ability to directly target key receptors and pathways that maintain central sensitization, making their effectiveness more pronounced. Although current drug therapy can alleviate NP, treatment satisfaction is still poor. The future development trajectory involves the creation of more targeted new drugs, specifically targeting the potential pathogenic mechanisms of central sensitization.

5.2 Biological Therapy

In recent years, biotherapy has emerged as a new strategy for the treatment of NP. Representative therapeutic drugs include monoclonal antibodies targeting anti-nerve growth factor (NGF) and anti-tumor necrosis factor (TNF) [152, 153]. NGF is involved in the process of peripheral and central pain sensitization. Monoclonal antibody targeting NGF can counteract the NGF-mediated pain amplification effect and show significant improvement in many clinical trials of NP [154, 155]. However, long-term use of these monoclonal antibodies can lead to adverse reactions, including bone and joint discomfort [156]. TNF-α is also an important inflammatory factor associated with NP. Anti-TNF monoclonal antibody can significantly reduce peripheral and central pain sensitivity [157, 158]. Compared with traditional analgesic drugs, biological agents have greater advantages in achieving therapeutic effects by accurately targeting key elements in the pain process. However, the long-term safety of some biological agents requires further evaluation. In the future, NP biotherapy is expected to include the development of new, safer and more effective biological agents for emerging central sensitization pathways.

5.3 Physiotherapy

The methods of relieving NP symptoms through physical therapy include transcutaneous electrical stimulation, low-power laser therapy, and spinal correction massage [159, 160, 161]. However, the effectiveness of these treatments is difficult to be confirmed. It is expected that precision medicine will bring more gratifying treatment results in the future. Exploring the mechanism of central sensitization and designing personalized new drugs are the key strategies to promote NP treatment in the future.

6. Conclusion and Prospect

In summary, NP is a common pain syndrome caused by nervous system injury, which seriously affects the quality of life of patients. The mechanism behind it is complicated. Recent studies have shown that central sensitization plays a key role in the pathogenesis and is associated with a series of molecular and cellular mechanisms. This includes the activation of glutamatergic excitatory transmission, the involvement of P2X4R and BDNF-TrkB signaling pathways, the enhancement of neuronal excitability and the weakening of inhibitory regulation. These changes ultimately lead to enhanced and amplified effects of central pain management. Although considerable progress has been made in studying the mechanism of central sensitization, it is still crucial to translate these findings into clinical applications. For example, new drug development targeting molecules such as P2X4R or BDNF is underway. Comprehensive evaluation requires large-scale randomized controlled trials to evaluate treatment efficacy and long-term safety. Although analgesic drug therapy can provide relief of some symptoms, challenges such as patient compliance significantly affect its effectiveness.

Looking ahead, our work will continue to focus on elucidating central sensitization mechanisms and identifying precise intervention points. This work will lead to the development of new targeted therapeutic drugs and ultimately improve the treatment level of NP. In the future, continuous drug screening efforts can continue, and the use of the latest technologies, such as single-cell genomics, to design inhibitors or antagonists targeting these key molecules is expected to produce more effective treatments. In addition, it is also imperative to improve the establishment of animal models to more accurately simulate clinical conditions and evaluate efficacy. In short, the study of central sensitization mechanism has far-reaching significance. Taking it as the cornerstone of designing new drug treatment strategies will promote the continuous development of NP treatment, and more researchers are expected to contribute in this field.

Author Contributions

WN and YM organized, designed, and wrote the article. ZK and YS designed of the work, drafted and revised this article for key intellectual content. WJ and WZ were responsible for literature collection and image creation. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We thank the Bone and Joint Research Institute and Cuiying Biological Center of the Second Hospital of Lanzhou University for their support.

Funding

This study was supported by the Fund Project of the Second Hospital of Lanzhou University (No. CY2022-QN-A03).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson AH, Yarnitsky D, et al. Neuropathic pain. Nature Reviews. Disease Primers. 2017; 3: 17002.
[2]
Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology. 2008; 70: 1630–1635.
[3]
Jensen TS, Baron R, Haanpää M, Kalso E, Loeser JD, Rice ASC, et al. A new definition of neuropathic pain. Pain. 2011; 152: 2204–2205.
[4]
van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain. 2014; 155: 654–662.
[5]
O’Connor AB, Dworkin RH. Treatment of neuropathic pain: an overview of recent guidelines. The American Journal of Medicine. 2009; 122: S22–S32.
[6]
Gilron I, Jensen TS, Dickenson AH. Combination pharmacotherapy for management of chronic pain: from bench to bedside. The Lancet. Neurology. 2013; 12: 1084–1095.
[7]
Schmelz M, Mantyh P, Malfait AM, Farrar J, Yaksh T, Tive L, et al. Nerve growth factor antibody for the treatment of osteoarthritis pain and chronic low-back pain: mechanism of action in the context of efficacy and safety. Pain. 2019; 160: 2210–2220.
[8]
Liu ZY, Song ZW, Guo SW, He JS, Wang SY, Zhu JG, et al. CXCL12/CXCR4 signaling contributes to neuropathic pain via central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neuroscience & Therapeutics. 2019; 25: 922–936.
[9]
Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. The Journal of Pain. 2009; 10: 895–926.
[10]
Ji RR, Nackley A, Huh Y, Terrando N, Maixner W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology. 2018; 129: 343–366.
[11]
Dou Y, Xia J, Gao R, Gao X, Munoz FM, Wei D, et al. Orai1 Plays a Crucial Role in Central Sensitization by Modulating Neuronal Excitability. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience. 2018; 38: 887–900.
[12]
Defaye M, Iftinca MC, Gadotti VM, Basso L, Abdullah NS, Cuménal M, et al. The neuronal tyrosine kinase receptor ligand ALKAL2 mediates persistent pain. The Journal of Clinical Investigation. 2022; 132: e154317.
[13]
Tinnirello A, Mazzoleni S, Santi C. Chronic Pain in the Elderly: Mechanisms and Distinctive Features. Biomolecules. 2021; 11: 1256.
[14]
Nijs J, Malfliet A, Ickmans K, Baert I, Meeus M. Treatment of central sensitization in patients with ‘unexplained’ chronic pain: an update. Expert Opinion on Pharmacotherapy. 2014; 15: 1671–1683.
[15]
Sariyildiz A, Coskun Benlidayi I, Turk I, Zengin Acemoglu SS, Unal I. Biopsychosocial factors should be considered when evaluating central sensitization in axial spondyloarthritis. Rheumatology International. 2023; 43: 923–932.
[16]
Guler MA, Celik OF, Ayhan FF. The important role of central sensitization in chronic musculoskeletal pain seen in different rheumatic diseases. Clinical Rheumatology. 2020; 39: 269–274.
[17]
Hansen MS, Asghar MS, Wetterslev J, Pipper CB, Mårtensson J, Becerra L, et al. The association between areas of secondary hyperalgesia and volumes of the caudate nuclei and other pain relevant brain structures-A 3-tesla MRI study of healthy men. PLoS ONE. 2018; 13: e0201642.
[18]
Souza Monteiro de Araujo D, Nassini R, Geppetti P, De Logu F. TRPA1 as a therapeutic target for nociceptive pain. Expert Opinion on Therapeutic Targets. 2020; 24: 997–1008.
[19]
Xie HT, Xia ZY, Pan X, Zhao B, Liu ZG. Puerarin ameliorates allodynia and hyperalgesia in rats with peripheral nerve injury. Neural Regeneration Research. 2018; 13: 1263–1268.
[20]
Pekošak A, Bulc JŽ, Korat Š, Schuit RC, Kooijman E, Vos R, et al. Synthesis and Preclinical Evaluation of the First Carbon-11 Labeled PET Tracers Targeting Substance P1-7. Molecular Pharmaceutics. 2018; 15: 4872–4883.
[21]
Zeng X, Mai J, Xie H, Yang L, Liu X. Activation of CB1R alleviates central sensitization by regulating HCN2-pNR2B signaling in a chronic migraine rat model. The Journal of Headache and Pain. 2023; 24: 44.
[22]
Drummond PD, Finch PM, Birklein F, Stanton-Hicks M, Knudsen LF. Hemisensory disturbances in patients with complex regional pain syndrome. Pain. 2018; 159: 1824–1832.
[23]
Kahle KT, Khanna A, Clapham DE, Woolf CJ. Therapeutic restoration of spinal inhibition via druggable enhancement of potassium-chloride cotransporter KCC2-mediated chloride extrusion in peripheral neuropathic pain. JAMA Neurology. 2014; 71: 640–645.
[24]
Zhao Y, Xin Y, Chu H. MC4R Is Involved in Neuropathic Pain by Regulating JNK Signaling Pathway After Chronic Constriction Injury. Frontiers in Neuroscience. 2019; 13: 919.
[25]
Lepore AC, O’Donnell J, Bonner JF, Paul C, Miller ME, Rauck B, et al. Spatial and temporal changes in promoter activity of the astrocyte glutamate transporter GLT1 following traumatic spinal cord injury. Journal of Neuroscience Research. 2011; 89: 1001–1017.
[26]
Milligan ED, Penzkover KR, Soderquist RG, Mahoney MJ. Spinal interleukin-10 therapy to treat peripheral neuropathic pain. Neuromodulation: Journal of the International Neuromodulation Society. 2012; 15: 520–526.
[27]
Dworkin RH, O’Connor AB, Kent J, Mackey SC, Raja SN, Stacey BR, et al. Interventional management of neuropathic pain: NeuPSIG recommendations. Pain. 2013; 154: 2249–2261.
[28]
Hasvik E, Haugen AJ, Grøvle L. Symptom descriptors and patterns in lumbar radicular pain caused by disc herniation: a 1-year longitudinal cohort study. BMJ Open. 2022; 12: e065500.
[29]
Brazill JM, Cruz B, Zhu Y, Zhai RG. Nmnat mitigates sensory dysfunction in a Drosophila model of paclitaxel-induced peripheral neuropathy. Disease Models & Mechanisms. 2018; 11: dmm032938.
[30]
Canneti A, Luzi M, Di Marco P, Cannata F, Pasqualitto F, Spinoglio A, et al. Safety and efficacy of transdermal buprenorphine and transdermal fentanyl in the treatment of neuropathic pain in AIDS patients. Minerva Anestesiologica. 2013; 79: 871–883.
[31]
Ji RR, Xu ZZ, Gao YJ. Emerging targets in neuroinflammation-driven chronic pain. Nature Reviews. Drug Discovery. 2014; 13: 533–548.
[32]
Fornasari D. Pain mechanisms in patients with chronic pain. Clinical Drug Investigation. 2012; 32: 45–52.
[33]
Zhang Z, Roberson DP, Kotoda M, Boivin B, Bohnslav JP, González-Cano R, et al. Automated preclinical detection of mechanical pain hypersensitivity and analgesia. Pain. 2022; 163: 2326–2336.
[34]
Jensen TS, Finnerup NB. Allodynia and hyperalgesia in neuropathic pain: clinical manifestations and mechanisms. The Lancet. Neurology. 2014; 13: 924–935.
[35]
Ando M, Hayashi Y, Hitomi S, Shibuta I, Furukawa A, Oto T, et al. Oxytocin-Dependent Regulation of TRPs Expression in Trigeminal Ganglion Neurons Attenuates Orofacial Neuropathic Pain Following Infraorbital Nerve Injury in Rats. International Journal of Molecular Sciences. 2020; 21: 9173.
[36]
Borsook D, Becerra L, Fishman S, Edwards A, Jennings CL, Stojanovic M, et al. Acute plasticity in the human somatosensory cortex following amputation. Neuroreport. 1998; 9: 1013–1017.
[37]
Lee PM, So Y, Park JM, Park CM, Kim HK, Kim JH. Spinal Cauda Equina Stimulation for Alternative Location of Spinal Cord Stimulation in Intractable Phantom Limb Pain Syndrome: A Case Report. The Korean Journal of Pain. 2016; 29: 123–128.
[38]
Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011; 152: S2–S15.
[39]
Lotze M, Moseley GL. Role of distorted body image in pain. Current Rheumatology Reports. 2007; 9: 488–496.
[40]
Wachholtz A, Foster S, Cheatle M. Psychophysiology of pain and opioid use: implications for managing pain in patients with an opioid use disorder. Drug and Alcohol Dependence. 2015; 146: 1–6.
[41]
Gwak YS, Hulsebosch CE, Leem JW. Neuronal-Glial Interactions Maintain Chronic Neuropathic Pain after Spinal Cord Injury. Neural Plasticity. 2017; 2017: 2480689.
[42]
Liu C, Zhang Y, Liu Q, Jiang L, Li M, Wang S, et al. P2X4-receptor participates in EAAT3 regulation via BDNF-TrkB signaling in a model of trigeminal allodynia. Molecular Pain. 2018; 14: 1744806918795930.
[43]
Zhao L, Li D, Liu N, Liu L, Zhang Z, Gao C, et al. Correlation of TGN-020 with the analgesic effects via ERK pathway activation after chronic constriction injury. Molecular Pain. 2018; 14: 1744806918796057.
[44]
Kuthati Y, Goutham Davuluri VN, Yang CP, Chang HC, Chang CP, Wong CS. Melatonin MT2 receptor agonist IIK-7 produces antinociception by modulation of ROS and suppression of spinal microglial activation in neuropathic pain rats. Journal of Pain Research. 2019; 12: 2473–2485.
[45]
Isami K, Haraguchi K, So K, Asakura K, Shirakawa H, Mori Y, et al. Involvement of TRPM2 in peripheral nerve injury-induced infiltration of peripheral immune cells into the spinal cord in mouse neuropathic pain model. PLoS ONE. 2013; 8: e66410.
[46]
Davenport AJ, Neagoe I, Bräuer N, Koch M, Rotgeri A, Nagel J, et al. Eliapixant is a selective P2X3 receptor antagonist for the treatment of disorders associated with hypersensitive nerve fibers. Scientific Reports. 2021; 11: 19877.
[47]
Liu JA, Yu J, Cheung CW. Immune Actions on the Peripheral Nervous System in Pain. International Journal of Molecular Sciences. 2021; 22: 1448.
[48]
Kocot-Kępska M, Zajączkowska R, Mika J, Wordliczek J, Dobrogowski J, Przeklasa-Muszyńska A. Peripheral Mechanisms of Neuropathic Pain-the Role of Neuronal and Non-Neuronal Interactions and Their Implications for Topical Treatment of Neuropathic Pain. Pharmaceuticals (Basel, Switzerland). 2021; 14: 77.
[49]
Ustaoglu A, Woodland P. Sensory Phenotype of the Oesophageal Mucosa in Gastro-Oesophageal Reflux Disease. International Journal of Molecular Sciences. 2023; 24: 2502.
[50]
Jiang Y, Wang J, Li H, Xia L. IL-35 alleviates inflammation progression in a rat model of diabetic neuropathic pain via inhibition of JNK signaling. Journal of Inflammation (London, England). 2019; 16: 19.
[51]
Tan CY, Wang YP, Han YY, Lu BH, Ji W, Zhu LC, et al. Expression and effect of sodium-potassium-chloride cotransporter on dorsal root ganglion neurons in a rat model of chronic constriction injury. Neural Regeneration Research. 2020; 15: 912–921.
[52]
Rekatsina M, Paladini A, Piroli A, Zis P, Pergolizzi JV, Varrassi G. Pathophysiologic Approach to Pain Therapy for Complex Pain Entities: A Narrative Review. Pain and Therapy. 2020; 9: 7–21.
[53]
Salat K, Gryzlo B, Kulig K. Experimental Drugs for Neuropathic Pain. Current Neuropharmacology. 2018; 16: 1193–1209.
[54]
Zhu D, Fan T, Huo X, Cui J, Cheung CW, Xia Z. Progressive Increase of Inflammatory CXCR4 and TNF-Alpha in the Dorsal Root Ganglia and Spinal Cord Maintains Peripheral and Central Sensitization to Diabetic Neuropathic Pain in Rats. Mediators of Inflammation. 2019; 2019: 4856156.
[55]
Kwok CHT, Learoyd AE, Canet-Pons J, Trang T, Fitzgerald M. Spinal interleukin-6 contributes to central sensitisation and persistent pain hypersensitivity in a model of juvenile idiopathic arthritis. Brain, Behavior, and Immunity. 2020; 90: 145–154.
[56]
Zhao Y, Zhu R, Xiao T, Liu X. Genetic variants in migraine: a field synopsis and systematic re-analysis of meta-analyses. The Journal of Headache and Pain. 2020; 21: 13.
[57]
Liu CC, Gao YJ, Luo H, Berta T, Xu ZZ, Ji RR, et al. Interferon alpha inhibits spinal cord synaptic and nociceptive transmission via neuronal-glial interactions. Scientific Reports. 2016; 6: 34356.
[58]
Zhao Y, Sharfman NM, Jaber VR, Lukiw WJ. Down-Regulation of Essential Synaptic Components by GI-Tract Microbiome-Derived Lipopolysaccharide (LPS) in LPS-Treated Human Neuronal-Glial (HNG) Cells in Primary Culture: Relevance to Alzheimer’s Disease (AD). Frontiers in Cellular Neuroscience. 2019; 13: 314.
[59]
Huang CY, Chen YL, Li AH, Lu JC, Wang HL. Minocycline, a microglial inhibitor, blocks spinal CCL2-induced heat hyperalgesia and augmentation of glutamatergic transmission in substantia gelatinosa neurons. Journal of Neuroinflammation. 2014; 11: 7.
[60]
Chen SR, Zhou HY, Byun HS, Chen H, Pan HL. Casein kinase II regulates N-methyl-D-aspartate receptor activity in spinal cords and pain hypersensitivity induced by nerve injury. The Journal of Pharmacology and Experimental Therapeutics. 2014; 350: 301–312.
[61]
Baron R, Baron Y, Disbrow E, Roberts TP. Activation of the somatosensory cortex during Abeta-fiber mediated hyperalgesia. A MSI study. Brain Research. 2000; 871: 75–82.
[62]
Ferrari LF, Lotufo CM, Araldi D, Rodrigues MA, Macedo LP, Ferreira SH, et al. Inflammatory sensitization of nociceptors depends on activation of NMDA receptors in DRG satellite cells. Proceedings of the National Academy of Sciences of the United States of America. 2014; 111: 18363–18368.
[63]
Johnston GA. GABAA receptor pharmacology. Pharmacology & Therapeutics. 1996; 69: 173–198.
[64]
Zhou HY, Chen SR, Pan HL. Targeting N-methyl-D-aspartate receptors for treatment of neuropathic pain. Expert Review of Clinical Pharmacology. 2011; 4: 379–388.
[65]
Chen YL, Feng XL, Cheung CW, Liu JA. Mode of action of astrocytes in pain: From the spinal cord to the brain. Progress in Neurobiology. 2022; 219: 102365.
[66]
Midavaine É, Côté J, Marchand S, Sarret P. Glial and neuroimmune cell choreography in sexually dimorphic pain signaling. Neuroscience and Biobehavioral Reviews. 2021; 125: 168–192.
[67]
Shkodra M, Brunelli C, Zecca E, Infante G, Miceli R, Caputo M, et al. Cancer pain: Results of a prospective study on prognostic indicators of pain intensity including pain syndromes assessment. Palliative Medicine. 2022; 36: 1396–1407.
[68]
Chau A, Steib S, Whitaker E, Kohns D, Quinter A, Craig A, et al. Theoretical Schemas to Guide Back Pain Consortium (BACPAC) Chronic Low Back Pain Clinical Research. Pain Medicine (Malden, Mass.). 2023; 24: S13–S35.
[69]
Warwick CA, Keyes AL, Woodruff TM, Usachev YM. The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. The Journal of Biological Chemistry. 2021; 297: 101085.
[70]
Yeh JF, Akinci A, Al Shaker M, Chang MH, Danilov A, Guileen R, et al. Monoclonal antibodies for chronic pain: a practical review of mechanisms and clinical applications. Molecular Pain. 2017; 13: 1744806917740233.
[71]
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug and Alcohol Dependence. 2017; 180: 156–170.
[72]
Zhao BS, Song XR, Hu PY, Meng LX, Tan YH, She YJ, et al. Hyperbaric oxygen treatment at various stages following chronic constriction injury produces different antinociceptive effects via regulation of P2X4R expression and apoptosis. PLoS ONE. 2015; 10: e0120122.
[73]
Caraci F, Merlo S, Drago F, Caruso G, Parenti C, Sortino MA. Rescue of Noradrenergic System as a Novel Pharmacological Strategy in the Treatment of Chronic Pain: Focus on Microglia Activation. Frontiers in Pharmacology. 2019; 10: 1024.
[74]
Ally A, Powell I, Ally MM, Chaitoff K, Nauli SM. Role of neuronal nitric oxide synthase on cardiovascular functions in physiological and pathophysiological states. Nitric Oxide: Biology and Chemistry. 2020; 102: 52–73.
[75]
Bathel A, Schweizer L, Stude P, Glaubitz B, Wulms N, Delice S, et al. Increased thalamic glutamate/glutamine levels in migraineurs. The Journal of Headache and Pain. 2018; 19: 55.
[76]
Kim S, Gang J, Lee JH, Yang H, Cheon C, Ko SG, et al. [6]-Shogaol Attenuates Oxaliplatin-Induced Allodynia through Serotonergic Receptors and GABA in the Spinal Cord in Mice. Pharmaceuticals. 2022; 15: 726.
[77]
Carlesso LC, Segal NA, Curtis JR, Wise BL, Frey Law L, Nevitt M, et al. Knee Pain and Structural Damage as Risk Factors for Incident Widespread Pain: Data From the Multicenter Osteoarthritis Study. Arthritis Care & Research. 2017; 69: 826–832.
[78]
Putatunda R, Hala TJ, Chin J, Lepore AC. Chronic at-level thermal hyperalgesia following rat cervical contusion spinal cord injury is accompanied by neuronal and astrocyte activation and loss of the astrocyte glutamate transporter, GLT1, in superficial dorsal horn. Brain Research. 2014; 1581: 64–79.
[79]
Wilsey B, Marcotte TD, Deutsch R, Zhao H, Prasad H, Phan A. An Exploratory Human Laboratory Experiment Evaluating Vaporized Cannabis in the Treatment of Neuropathic Pain From Spinal Cord Injury and Disease. The Journal of Pain. 2016; 17: 982–1000.
[80]
Deng L, Guindon J, Vemuri VK, Thakur GA, White FA, Makriyannis A, et al. The maintenance of cisplatin- and paclitaxel-induced mechanical and cold allodynia is suppressed by cannabinoid CB₂ receptor activation and independent of CXCR4 signaling in models of chemotherapy-induced peripheral neuropathy. Molecular Pain. 2012; 8: 71.
[81]
Lai A, Iliff D, Zaheer K, Wang D, Gansau J, Laudier DM, et al. Spinal Cord Sensitization and Spinal Inflammation from an In Vivo Rat Endplate Injury Associated with Painful Intervertebral Disc Degeneration. International Journal of Molecular Sciences. 2023; 24: 3425.
[82]
Webber CA, Salame J, Luu GLS, Acharjee S, Ruangkittisakul A, Martinez JA, et al. Nerve growth factor acts through the TrkA receptor to protect sensory neurons from the damaging effects of the HIV-1 viral protein, Vpr. Neuroscience. 2013; 252: 512–525.
[83]
Lee JHA, Chen Q, Zhuo M. Synaptic Plasticity in the Pain-Related Cingulate and Insular Cortex. Biomedicines. 2022; 10: 2745.
[84]
Chen JL, Zhou X, Liu BL, Wei XH, Ding HL, Lin ZJ, et al. Normalization of magnesium deficiency attenuated mechanical allodynia, depressive-like behaviors, and memory deficits associated with cyclophosphamide-induced cystitis by inhibiting TNF-α/NF-κB signaling in female rats. Journal of Neuroinflammation. 2020; 17: 99.
[85]
Dellarole A, Morton P, Brambilla R, Walters W, Summers S, Bernardes D, et al. Neuropathic pain-induced depressive-like behavior and hippocampal neurogenesis and plasticity are dependent on TNFR1 signaling. Brain, Behavior, and Immunity. 2014; 41: 65–81.
[86]
Jensen KB, Loitoile R, Kosek E, Petzke F, Carville S, Fransson P, et al. Patients with fibromyalgia display less functional connectivity in the brain’s pain inhibitory network. Molecular Pain. 2012; 8: 32.
[87]
Long T, He W, Pan Q, Zhang S, Zhang D, Qin G, et al. Microglia P2X4R-BDNF signalling contributes to central sensitization in a recurrent nitroglycerin-induced chronic migraine model. The Journal of Headache and Pain. 2020; 21: 4.
[88]
Goncalves MB, Moehlin J, Clarke E, Grist J, Hobbs C, Carr AM, et al. RARβ Agonist Drug (C286) Demonstrates Efficacy in a Pre-clinical Neuropathic Pain Model Restoring Multiple Pathways via DNA Repair Mechanisms. iScience. 2019; 20: 554–566.
[89]
Zhou WM, Lei ZY, Shi YQ, Gong CY, Kai Z, Wei N, et al. Intrathecal Injection of Botulinum Toxin Type A has an Analgesic Effect in Male Rats CCI Model by Inhibiting the Activation of Spinal P2X4R. Neurochemical Research. 2023; 48: 3099–3112.
[90]
Mahdian Dehkordi F, Kaboutari J, Zendehdel M, Javdani M. The antinociceptive effect of artemisinin on the inflammatory pain and role of GABAergic and opioidergic systems. The Korean Journal of Pain. 2019; 32: 160–167.
[91]
Trapero C, Martín-Satué M. Purinergic Signaling in Endometriosis-Associated Pain. International Journal of Molecular Sciences. 2020; 21: 8512.
[92]
Lueptow LM, Fakira AK, Bobeck EN. The Contribution of the Descending Pain Modulatory Pathway in Opioid Tolerance. Frontiers in Neuroscience. 2018; 12: 886.
[93]
Ducza L, Gajtkó A, Hegedűs K, Bakk E, Kis G, Gaál B, et al. Neuronal P2X4 receptor may contribute to peripheral inflammatory pain in rat spinal dorsal horn. Frontiers in Molecular Neuroscience. 2023; 16: 1115685.
[94]
Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, et al. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003; 424: 778–783.
[95]
Coull JAM, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, et al. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005; 438: 1017–1021.
[96]
Beggs S, Trang T, Salter MW. P2X4R+ microglia drive neuropathic pain. Nature Neuroscience. 2012; 15: 1068–1073.
[97]
Silva GD, Lopes PSS, Fonoff ET, Pagano RL. The spinal anti-inflammatory mechanism of motor cortex stimulation: cause of success and refractoriness in neuropathic pain? Journal of Neuroinflammation. 2015; 12: 10.
[98]
Tang Y, Chen Y, Yang M, Zheng Q, Li Y, Bao Y. Knockdown of PAR2 alleviates cancer-induced bone pain by inhibiting the activation of astrocytes and the ERK pathway. BMC Musculoskeletal Disorders. 2022; 23: 514.
[99]
Tsuda M, Tozaki-Saitoh H, Masuda T, Toyomitsu E, Tezuka T, Yamamoto T, et al. Lyn tyrosine kinase is required for P2X(4) receptor upregulation and neuropathic pain after peripheral nerve injury. Glia. 2008; 56: 50–58.
[100]
Reyna-Jeldes M, De la Fuente-Ortega E, Cerda D, Velázquez-Miranda E, Pinto K, Vázquez-Cuevas FG, et al. Purinergic P2Y2 and P2X4 Receptors Are Involved in the Epithelial-Mesenchymal Transition and Metastatic Potential of Gastric Cancer Derived Cell Lines. Pharmaceutics. 2021; 13: 1234.
[101]
Robilotto GL, Mohapatra DP, Shepherd AJ, Mickle AD. Role of Src kinase in regulating protein kinase C mediated phosphorylation of TRPV1. European Journal of Pain (London, England). 2022; 26: 1967–1978.
[102]
Zúñiga-Romero Á, Rivera-Plata Q, Arrieta J, Flores-Murrieta FJ, Rodríguez-Silverio J, Reyes-García JG, et al. GPR55 and GPR119 Receptors Contribute to the Processing of Neuropathic Pain in Rats. Pharmaceuticals (Basel, Switzerland). 2022; 15: 67.
[103]
Zong W, Lu X, Dong G, Zhang L, Li K. Molecular mechanisms of exercise intervention in alleviating the symptoms of autism spectrum disorder: Targeting the structural alterations of synapse. Frontiers in Psychiatry. 2023; 14: 1096503.
[104]
Lee JM, Choi YJ, Yoo MC, Yeo SG. Central Facial Nervous System Biomolecules Involved in Peripheral Facial Nerve Injury Responses and Potential Therapeutic Strategies. Antioxidants (Basel, Switzerland). 2023; 12: 1036.
[105]
Huang YJ, Lee KH, Grau JW. Complete spinal cord injury (SCI) transforms how brain derived neurotrophic factor (BDNF) affects nociceptive sensitization. Experimental Neurology. 2017; 288: 38–50.
[106]
Wang Y, Mei X, Zhang L, Lv G. The correlation among the dynamic change of Zn2+, ZnT-1, and brain-derived neurotrophic factor after acute spinal cord injury in rats. Biological Trace Element Research. 2011; 143: 351–358.
[107]
Falnikar A, Hala TJ, Poulsen DJ, Lepore AC. GLT1 overexpression reverses established neuropathic pain-related behavior and attenuates chronic dorsal horn neuron activation following cervical spinal cord injury. Glia. 2016; 64: 396–406.
[108]
Geng SJ, Liao FF, Dang WH, Ding X, Liu XD, Cai J, et al. Contribution of the spinal cord BDNF to the development of neuropathic pain by activation of the NR2B-containing NMDA receptors in rats with spinal nerve ligation. Experimental Neurology. 2010; 222: 256–266.
[109]
Li J, Wang X, Wang H, Wang R, Guo Y, Xu L, et al. The BDNF-TrkB signaling pathway in the rostral anterior cingulate cortex is involved in the development of pain aversion in rats with bone cancer via NR2B and ERK-CREB signaling. Brain Research Bulletin. 2022; 185: 18–27.
[110]
Infantino R, Schiano C, Luongo L, Paino S, Mansueto G, Boccella S, et al. MED1/BDNF/TrkB pathway is involved in thalamic hemorrhage-induced pain and depression by regulating microglia. Neurobiology of Disease. 2022; 164: 105611.
[111]
Wang ZH, Xiang J, Liu X, Yu SP, Manfredsson FP, Sandoval IM, et al. Deficiency in BDNF/TrkB Neurotrophic Activity Stimulates δ-Secretase by Upregulating C/EBPβ in Alzheimer’s Disease. Cell Reports. 2019; 28: 655–669.e5.
[112]
Romaus-Sanjurjo D, Rodicio MC, Barreiro-Iglesias A. Gamma-aminobutyric acid (GABA) promotes recovery from spinal cord injury in lampreys: role of GABA receptors and perspective on the translation to mammals. Neural Regeneration Research. 2019; 14: 1695–1696.
[113]
Zunszain PA, Horowitz MA, Cattaneo A, Lupi MM, Pariante CM. Ketamine: synaptogenesis, immunomodulation and glycogen synthase kinase-3 as underlying mechanisms of its antidepressant properties. Molecular Psychiatry. 2013; 18: 1236–1241.
[114]
Cataldo G, Rajput S, Gupta K, Simone DA. Sensitization of nociceptive spinal neurons contributes to pain in a transgenic model of sickle cell disease. Pain. 2015; 156: 722–730.
[115]
Ji RR. Neuroimmune interactions in itch: Do chronic itch, chronic pain, and chronic cough share similar mechanisms? Pulmonary Pharmacology & Therapeutics. 2015; 35: 81–86.
[116]
Takasu K, Sakai A, Hanawa H, Shimada T, Suzuki H. Overexpression of GDNF in the uninjured DRG exerts analgesic effects on neuropathic pain following segmental spinal nerve ligation in mice. The Journal of Pain. 2011; 12: 1130–1139.
[117]
Carter MW, Johnson KM, Lee JY, Hulsebosch CE, Gwak YS. Comparison of Mechanical Allodynia and Recovery of Locomotion and Bladder Function by Different Parameters of Low Thoracic Spinal Contusion Injury in Rats. The Korean Journal of Pain. 2016; 29: 86–95.
[118]
Palazzo E, Marabese I, Gargano F, Guida F, Belardo C, Maione S. Methods for Evaluating Sensory, Affective and Cognitive Disorders in Neuropathic Rodents. Current Neuropharmacology. 2021; 19: 736–746.
[119]
Li J, Stratton HJ, Lorca SA, Grace PM, Khanna R. Small molecule targeting NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in chronic constriction injury (CCI) rats. Channels (Austin, Tex.). 2022; 16: 1–8.
[120]
Zhi MJ, Liu K, Zheng ZL, He X, Li T, Sun G, et al. Application of the chronic constriction injury of the partial sciatic nerve model to assess acupuncture analgesia. Journal of Pain Research. 2017; 10: 2271–2280.
[121]
Bennett GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain. 1988; 33: 87–107.
[122]
Challa SR. Surgical animal models of neuropathic pain: Pros and Cons. The International Journal of Neuroscience. 2015; 125: 170–174.
[123]
Vincenzetti S, Pucciarelli S, Huang Y, Ricciutelli M, Lambertucci C, Volpini R, et al. Biomarkers mapping of neuropathic pain in a nerve chronic constriction injury mice model. Biochimie. 2019; 158: 172–179.
[124]
Seltzer Z, Dubner R, Shir Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain. 1990; 43: 205–218.
[125]
Ho Kim S, Mo Chung J. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain. 1992; 50: 355–363.
[126]
Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000; 87: 149–158.
[127]
Bourquin AF, Süveges M, Pertin M, Gilliard N, Sardy S, Davison AC, et al. Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse. Pain. 2006; 122: 14.e1-14.
[128]
Li L, Xian CJ, Zhong JH, Zhou XF. Effect of lumbar 5 ventral root transection on pain behaviors: a novel rat model for neuropathic pain without axotomy of primary sensory neurons. Experimental Neurology. 2002; 175: 23–34.
[129]
Hu SJ, Xing JL. An experimental model for chronic compression of dorsal root ganglion produced by intervertebral foramen stenosis in the rat. Pain. 1998; 77: 15–23.
[130]
Lee MC, Nam TS, Jung SJ, Gwak YS, Leem JW. Modulation of Spinal GABAergic Inhibition and Mechanical Hypersensitivity following Chronic Compression of Dorsal Root Ganglion in the Rat. Neural Plasticity. 2015; 2015: 924728.
[131]
Shiao R, Lee-Kubli CA. Neuropathic Pain After Spinal Cord Injury: Challenges and Research Perspectives. Neurotherapeutics: the Journal of the American Society for Experimental NeuroTherapeutics. 2018; 15: 635–653.
[132]
Mogil JS. Animal models of pain: progress and challenges. Nature Reviews. Neuroscience. 2009; 10: 283–294.
[133]
Zhang H, Xie W, Xie Y. Spinal cord injury triggers sensitization of wide dynamic range dorsal horn neurons in segments rostral to the injury. Brain Research. 2005; 1055: 103–110.
[134]
Hanada T, Kurihara T, Tokudome M, Tokimura H, Arita K, Miyata A. Development and pharmacological verification of a new mouse model of central post-stroke pain. Neuroscience Research. 2014; 78: 72–80.
[135]
Gritsch S, Bali KK, Kuner R, Vardeh D. Functional characterization of a mouse model for central post-stroke pain. Molecular Pain. 2016; 12: 1744806916629049.
[136]
Cruccu G, Truini A, Neuropathic Pain Special Interest Group of the Italian Society of Neurology (Italian NeuPSIG). Neuropathic Pain: The Scope of the Problem. Pain and Therapy. 2017; 6: 1–3.
[137]
Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. The Lancet. Neurology. 2015; 14: 162–173.
[138]
Yalcin I, Choucair-Jaafar N, Benbouzid M, Tessier LH, Muller A, Hein L, et al. beta(2)-adrenoceptors are critical for antidepressant treatment of neuropathic pain. Annals of Neurology. 2009; 65: 218–225.
[139]
Obata H. Analgesic Mechanisms of Antidepressants for Neuropathic Pain. International Journal of Molecular Sciences. 2017; 18: 2483.
[140]
Huang C, Li WG, Zhang XB, Wang L, Xu TL, Wu D, et al. α-asarone from Acorus gramineus alleviates epilepsy by modulating A-type GABA receptors. Neuropharmacology. 2013; 65: 1–11.
[141]
Koo BS, Park KS, Ha JH, Park JH, Lim JC, Lee DU. Inhibitory effects of the fragrance inhalation of essential oil from Acorus gramineus on central nervous system. Biological & Pharmaceutical Bulletin. 2003; 26: 978–982.
[142]
Jordan B, Devi LA. Molecular mechanisms of opioid receptor signal transduction. British Journal of Anaesthesia. 1998; 81: 12–19.
[143]
Sarne Y, Fields A, Keren O, Gafni M. Stimulatory effects of opioids on transmitter release and possible cellular mechanisms: overview and original results. Neurochemical Research. 1996; 21: 1353–1361.
[144]
Saito J, Zao H, Wu L, Iwasaki M, Sun Q, Hu C, et al. ”Anti-cancer” effect of ketamine in comparison with MK801 on neuroglioma and lung cancer cells. European Journal of Pharmacology. 2023; 945: 175580.
[145]
Inoue K. The function of microglia through purinergic receptors: neuropathic pain and cytokine release. Pharmacology & Therapeutics. 2006; 109: 210–226.
[146]
Joshi HP, Jo HJ, Kim YH, An SB, Park CK, Han I. Stem Cell Therapy for Modulating Neuroinflammation in Neuropathic Pain. International Journal of Molecular Sciences. 2021; 22: 4853.
[147]
St John Smith E. Advances in understanding nociception and neuropathic pain. Journal of Neurology. 2018; 265: 231–238.
[148]
Zima AV, Qin J, Fill M, Blatter LA. Tricyclic antidepressant amitriptyline alters sarcoplasmic reticulum calcium handling in ventricular myocytes. American Journal of Physiology. Heart and Circulatory Physiology. 2008; 295: H2008–H2016.
[149]
Enke O, New HA, New CH, Mathieson S, McLachlan AJ, Latimer J, et al. Anticonvulsants in the treatment of low back pain and lumbar radicular pain: a systematic review and meta-analysis. CMAJ: Canadian Medical Association Journal = Journal De L’Association Medicale Canadienne. 2018; 190: E786–E793.
[150]
Kalso E, Edwards JE, Moore AR, McQuay HJ. Opioids in chronic non-cancer pain: systematic review of efficacy and safety. Pain. 2004; 112: 372–380.
[151]
Eccleston C, Fisher E, Thomas KH, Hearn L, Derry S, Stannard C, et al. Interventions for the reduction of prescribed opioid use in chronic non-cancer pain. The Cochrane Database of Systematic Reviews. 2017; 11: CD010323.
[152]
Liang ZJ, Tan J, Tang L, Xie ZB, Chen GJ, Liu GJ, et al. NGF monoclonal antibody DS002 alleviates chemotherapy-induced peripheral neuropathy in rats. Acta Pharmacologica Sinica. 2022; 43: 2841–2847.
[153]
Chiorazzi A, Canta A, Meregalli C, Carozzi V, Sala B, Oggioni N, et al. Antibody against tumor necrosis factor-α reduces bortezomib-induced allodynia in a rat model. Anticancer Research. 2013; 33: 5453–5459.
[154]
Zhao D, Zeng LF, Liang GH, Pan JK, Luo MH, Han YH, et al. Does anti-nerve growth factor monoclonal antibody treatment have the potential to replace nonsteroidal anti-inflammatory drugs and opioids in treating hip or knee osteoarthritis? A systematic review of randomized controlled trials. EFORT Open Reviews. 2022; 7: 470–480.
[155]
Enomoto M, Mantyh PW, Murrell J, Innes JF, Lascelles BDX. Anti-nerve growth factor monoclonal antibodies for the control of pain in dogs and cats. The Veterinary Record. 2019; 184: 23.
[156]
Roemer FW, Guermazi A. Imaging atlas for eligibility and on-study safety of potential joint adverse events in anti-NGF studies. Osteoarthritis and Cartilage. 2015; 23: S1–S2.
[157]
Thalayasingam N, Isaacs JD. Anti-TNF therapy. Best Practice & Research. Clinical Rheumatology. 2011; 25: 549–567.
[158]
Dirckx M, Groeneweg G, Wesseldijk F, Stronks DL, Huygen FJPM. Report of a preliminary discontinued double-blind, randomized, placebo-controlled trial of the anti-TNF-α chimeric monoclonal antibody infliximab in complex regional pain syndrome. Pain Practice: the Official Journal of World Institute of Pain. 2013; 13: 633–640.
[159]
Ramachandran VS, Rogers-Ramachandran D. Synaesthesia in phantom limbs induced with mirrors. Proceedings. Biological Sciences. 1996; 263: 377–386.
[160]
Whipple RR, Unsell RS. Treatment of painful neuromas. The Orthopedic Clinics of North America. 1988; 19: 175–185.
[161]
Ducic I, Mesbahi AN, Attinger CE, Graw K. The role of peripheral nerve surgery in the treatment of chronic pain associated with amputation stumps. Plastic and Reconstructive Surgery. 2008; 121: 908–914.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top