Skip to content
Publicly Available Published by De Gruyter November 10, 2022

An update to pain management after spinal cord injury: from pharmacology to circRNAs

  • Graciela L. Mazzone ORCID logo EMAIL logo , María F. Coronel ORCID logo , Miranda Mladinic ORCID logo and Cynthia Sámano ORCID logo EMAIL logo

Abstract

Neuropathic pain (NP) following a spinal cord injury (SCI) is often hard to control and therapies should be focused on the physical, psychological, behavioral, social, and environmental factors that may contribute to chronic sensory symptoms. Novel therapeutic treatments for NP management should be based on the combination of pharmacological and nonpharmacological options. Some of them are addressed in this review with a focus on mechanisms and novel treatments. Several reports demonstrated an aberrant expression of non-coding RNAs (ncRNAs) that may represent key regulatory factors with a crucial role in the pathophysiology of NP and as potential diagnostic biomarkers. This review analyses the latest evidence for cellular and molecular mechanisms associated with the role of circular RNAs (circRNAs) in the management of pain after SCI. Advantages in the use of circRNA are their stability (up to 48 h), and specificity as sponges of different miRNAs related to SCI and nerve injury. The present review discusses novel data about deregulated circRNAs (up or downregulated) that sponge miRNAs, and promote cellular and molecular interactions with mRNAs and proteins. This data support the concept that circRNAs could be considered as novel potential therapeutic targets for NP management especially after spinal cord injuries.

Introduction: origin and classification of pain after SCI

The estimated annual incidence for spinal cord injury (SCI) is 15–40 cases per million, mostly affecting young adults with severe and permanent disabilities (Berg et al. 2011; Johansson et al. 2021; Miyakoshi et al. 2021; Witiw and Fehlings 2015). The pathophysiology of SCI involves primary and secondary mechanisms that affect sensory, motor, and autonomic function below the level of injury (Witiw and Fehlings 2015). The primary injury includes physical and mechanical trauma, contusion, compression, and concussion, which cause local, segmental, and limited damage to the spinal cord, generating breakage or contusion of axons, hemorrhage, ischemia, and edema (Ahuja et al. 2017; Park et al. 2004; Witiw and Fehlings 2015). The later events after a primary injury involve a complex cascade of degenerative processes termed secondary injury. Temporally, the secondary damage considerably amplifies the destruction of neuronal and glial cells in areas adjacent to the primary injury (Brockie et al. 2021; Park et al. 2004) mainly by excitotoxicity process due to excessive release of glutamate. Pain is a consequence and frequent complication of SCI (Burke et al. 2017; Finnerup et al. 2001). After SCI, pain also referred to as “central pain” is related to emotional discomfort and limitation of physical activity, participation in work and social events, thus deteriorating the quality of life of the patient (Burke et al. 2017; Finnerup 2013; Hassanpour et al. 2012; Masri and Keller 2012). Central pain initiated by SCI manifests in diverse ways at any level of the spinal cord (Hunt et al. 2021; Wang et al. 2021c). The International Spinal Cord Injury Pain (ISCIP) designed a classification of pain directly related to SCI: nociceptive, neuropathic, other or unknown pain, which depend on the source and/or pathology (Bryce et al. 2012a, b). In particular, neuropathic pain (NP) often accompanies the functional deficits associated with SCI and is manifested with chronic sensory symptoms and signs (Burke et al. 2017). With this classification pain is defined as nociceptive (musculoskeletal or visceral), neuropathic (at-level or below-level of the spinal injury) or undefined pain (neither nociceptive nor neuropathic) (Latremoliere and Woolf 2009; Widerström-Noga 2017). Although there are distinct definitions of NP it is widely accepted that it is caused by a lesion or disease of the somatosensory system. Indeed, patients suffering from NP present spontaneous persistent pain and a variety of abnormally evoked responses to innocuous stimuli (allodynia) or increased response to noxious stimuli (hyperalgesia). Nowadays, SCI patients often develop NP that is refractory to treatment and is associated with adverse effects or withdrawal reactions that limit drug usefulness (Colloca et al. 2017). Thus, caution must be used when selecting treatment (Thomas et al. 2022). Indeed, clinical trials approved by the FDA are devised to reduce the toxicity and addictive potential of steroidal anti-inflammatory drugs employed for SCI (Mazaleuskaya et al. 2021).

Novel pharmacological interventions to treat NP after SCI

The most frequently used drugs include pregabalin, gabapentin, duloxetine (serotonin-noradrenaline reuptake inhibitor), various tricyclic antidepressants, capsaicin (the active component of chili peppers), lidocaine, and weak or strong opioids (tramadol) (Colloca et al. 2017; Wiffen et al. 2017). However, several clinical studies now point to the importance of achieving safe and efficacious treatment of pain by reducing the side effects of opioid prescription (Britch and Walsh 2022; Mazaleuskaya et al. 2021). Indeed, the use of natural compounds, such as glycosides (related to sugar metabolism) and resveratrol (stilbenoid polyphenol) obtained from medical plants have potential use for NP treatment (Miguel et al. 2021; Tian et al. 2021).

Antisense oligonucleotides are an increasingly represented class of drugs used to modulate expression levels by targeting both coding and non-coding RNA and are under FDA approval for genetic diseases, cancer, and neurodegenerative diseases (Quemener et al. 2022). On the other hand, immunomodulatory oligodeoxynucleotides (ODNs) are synthetic molecules that influence different types of cells of the vertebrate immune system. Indeed, IMT504, a prototype of the PyNTTTTGT family of ODNs (Coronel et al. 2008; Elias et al. 2003) has been shown to have promising translational results given its low toxicity and its beneficial effects on animal models of neuropathic pain and diabetes (Bianchi et al. 2012; Casadei et al. 2022; Coronel et al. 2008; Leiguarda et al. 2018, 2021a, 2021b). It has also been reported that the systematic application of IMT504 induces the mobilisation of mesenchymal stem cells to the peripheral blood, a phenomenon which could be involved in a modulation of the inflammatory response by molecular mechanisms that remain to be established (Zorzopulos et al. 2017). The regulation of the inflammatory response and its molecular mechanism could be a potential therapeutic strategy to improve prognosis and pain modulation after SCI. Unfortunately, NP after SCI is still difficult to treat, largely because the underlying mechanisms are not yet fully understood. Thus, with a better understanding of the molecular mechanisms, optimal approaches to manage NP can be achieved which in turn could be crucial to develop novel diagnostic and treatment tools.

Overview of molecular modulators during evolvement and progression of neuropathic pain

NP after SCI is complex and develops as a consequence of a cascade of molecular events that lead to plastic changes that contribute to the development and maintenance of pain and remain incompletely understood. The pathophysiology of NP principally affects sensory fibers (Aβ, Aδ and C fibers) and involves many signaling cascades (Colloca et al. 2017; Finnerup et al. 2001; Nikolenko et al. 2022), such as neuroinflammatory response to injury, ectopic action potential discharge, upregulation of chemokines and chemokine receptors (Knerlich-Lukoschus et al. 2011), changes in calcium channel expression (Boroujerdi et al. 2011; Park and Luo 2010), modulation of expression and function of N-Methyl-D-Aspartate (NMDA) (Qu et al. 2009) and purine receptors, or opioids metabolism (Burnstock 2020; Chen et al. 2012; Detloff et al. 2014; Wang et al. 2021b), and brain-derived neurotrophic factor (BDNF) (Geng et al. 2010; Qu et al. 2009). After SCI there is also an alteration in the gain of excitation and the modulation of the inhibition with a significant role for extrasynaptic GABAA receptors in sparing spinal cord neurons from injury (Mazzone and Nistri 2019; Mazzone et al. 2021). Indeed, the changes in sensory pathways towards hyperexcitability over time might contribute to the NP becoming chronic (Colloca et al. 2017; Finnerup et al. 2021). Similarly, several reports have analysed the basis of cellular targets and circuits for persistent pain conditions, that are initiated by dysregulation of primary sensory neurons in the spinal cord network and brain, to explain peripheral plasticity in pathological disease state (Kuner 2010; Prescott et al. 2014; von Hehn et al. 2012). Indeed, the communication between brain and periphery, which occurs bidirectionally through reciprocal autonomic nervous system mechanisms, hormones and humoral factors, is regulated by the cholinergic anti-inflammatory pathway (Pavlov and Tracey 2005) and it is an important regulatory point for inflammation-related pain diseases with numerous therapeutic potential currently under clinical trials (Kelly et al. 2022). In this line it was recently demonstrated in 49 female patients with fibromyalgia syndrome that 19 microRNA regulators of cholinergic transcripts, such as has-miR-27b-3p, has-miR-148a-3p, and has-miR-182-5p, are dysregulated in support of the theory that microRNA control immune cell processes (Erbacher et al. 2022).

On the other hand, emerging evidence has shown that the dorsal horn neurons of the spinal cord contribute to pain-like hypersensitivity through the transitory expression of VGLUT3 for mechanical allodynia (Peirs et al. 2015). The immune responses in the dorsal horn have been implicated in SCI-pain, but immune mechanisms in the periphery, especially in the dorsal root ganglia (DRG), where nociceptor cell bodies reside, have not been elucidated. Chronic pain (such as inflammatory and NP) is believed to be caused by aberrant neuronal responses along the pain transmission pathway from peripheral spinal cord, thalamus and cortex (Zhuo 2007). Besides that, sensory neuron-associated macrophages (sNAMs) have been involved in the pathophysiology of neuropathic pain via proinflammatory and pronociceptive mediators (Yu et al. 2020). The activation of sNAMs after a systemic injury has been suggested to stimulate the immune receptors Toll-like receptors (TLRs) (Lacagnina et al. 2018; Liu et al. 2012), which induce proinflammatory factors such as chemokines/cytokines (Yu et al. 2020). In addition, the cytoplasmic nucleotide-binding oligomerization domain-like receptors (NLRs), such as nucleotide-binding oligomerization domain 2 (NOD2) commonly expressed in the microglial cells, have been found to be upregulated in sNAMs of the sensory ganglia after peripheral nerve injury. The stimulation of NLRs may recruit receptor-interacting serine/threonine-protein kinase 2 (Santa-Cecília et al. 2019), which in turn activates the nuclear transcription factor kappa B regulating the transcription of proinflammatory genes (Hasegawa et al. 2008; Santa-Cecília et al. 2019), suggesting that TLRs and NLRs may play an important role in the development of NP.

Neurons and glial cells (astrocytes and microglia) have close interactions regulating the initiation and maintenance of NP (Crown 2012; Tsuda 2016), in fact, the time course of astrocyte activation has been shown to coincide with the transition from the acute to chronic states during NP (Donnelly et al. 2020; Li et al. 2019). In this context, it has been reported that chronic inflammation-induced hyperalgesia and NP are associated with reduced levels of serine–threonine kinase G-protein receptor kinase 2 (GRK 2) in spinal microglia/macrophage (Willemen et al. 2010). Recently, it has been reported in a mouse model of inflammatory pain that the neuronal spinal GRK2 mediates microglial activation and neuroinflammation (Chen et al. 2022b). Other factors, such as cell-surface receptors for neurotransmission, the mitogen-activated protein kinases, the extracellular signal-regulated protein kinase and c-Jun N-terminal kinase (MAPKs, ERK, and JNK), the proinflammatory cytokines and neurotrophic factors expression (Chen et al. 2022b; Crown 2012; Kobayashi et al. 2008; Willemen et al. 2010; Zhuang et al. 2005), have been also shown to promote NP through the activation of spinal microglia.

In this sense, studies based on electroacupuncture (EA) treatment have demonstrated that alleviation of spinal nerve ligation (SNL)-induced NP is promoted by inhibiting spinal microglial and astrocyte activation (Liang et al. 2016). The central mechanism of EA-induced analgesia has been partially attributed to the reduction of the expression of p-38 MAPK on microglia (Liang et al. 2016). Following this line, a recent study has reported that EA treatment alleviates NP by decreasing abnormal dendritic spine/synaptic reconstruction and inflammation via P2X7 receptor (P2X7R) downregulation during activation in microglia during the development of NP (Wu et al. 2021). In addition, the CXCL12/CXCR4 signaling pathway expression has also been found to be activated in chronic post-ischemia pain models. CXCL12 is mainly expressed in neurons and microglia while CXCR4 is both in neurons and astrocytes as important regulator and contributor to the development and maintenance of NP via peripheral and central sensitization mechanisms (Dubový et al. 2010; Liu et al. 2019; Luo et al. 2016; Shen et al. 2014). Interestingly, some investigations have also suggested that the regulation of pathological pain would differ between sexes (Chen et al. 2018), due to cellular and molecular signaling pathways induced by sex-hormones fluctuations in females. All these data support the concept that the inflammatory processes may contribute to develop and maintain NP induced by chronic injury of the sciatic nerve model in mice (Berta et al. 2016; Chen et al. 2018) and suggest the necessity of exploring several the aspects of pain-processing which can be sex dependent.

Other potentially important contributors to physiological and pathological pain are the metabotropic glutamate receptors (mGluR) and glycine transporters such as GlyT1 (Lu et al. 2022; Napier et al. 2012; Tao et al. 2005). Development of selective mGluR antagonists has demonstrated the involvement of the metabotropic receptors in chronic pain modulation (Goudet et al. 2008; Palazzo et al. 2017). In SNL injury models mechanical allodynia is ameliorated by an inhibitor of GlyT1 and by GlyT1 knockdown (Morita et al. 2008). More recently, data provided by Lu et al. demonstrate that the intrathecal anesthetic bupivacaine reduces GlyT1 expression in spinal astrocytes by activating the p-AMPK/BDNF signaling pathway, suggesting that regulation of GlyT1 expression could be a crucial mechanism to modulate the NP mechanisms (Lu et al. 2022). Although glutamate-receptor antagonists have shown effectiveness in reducing pathological pain in animal models, their usefulness in clinical trials has been limited, probably due to their side effects (Collins et al. 2010; Hewitt 2000). Thus, the main targets for effective pharmacological intervention on NP are considered the neuronal circuitry activity and the immune and inflammatory cells. Thus, the main targets for effective pharmacological intervention of NP are considered to be the neuronal circuitry activity and the immune and inflammatory cells (Nakagawa and Kaneko 2010). However, NP is a complex condition and therefore the pharmacological plan should be based on each patient´s clinical history (Jongen et al. 2014; Wu et al. 2021; Zhuo et al. 2011). In this sense, evidence has shown the possible role of non-coding RNAs in modulating the development and progression of NP through the functional expression of several members of transient receptor potential (TRP) and voltage-gated ion (Na+, Ca2+, K+, and ClV) channels (Felix et al. 2022). Collectively, all these data suggest the need to identify the molecular pathways and cellular targets as novel potential effectors involved in chronic pain treatment (Chen et al. 2022b; Willemen et al. 2010). In the next section, we will address the novel players in NP management related to non-coding RNAs from a cellular to molecular point of view.

The role of non-coding RNAs in pain pathological mechanisms: special focus on circular RNAs

Non-coding RNAs (ncRNAs) are a class of RNA molecules that typically do not encode proteins but functionally regulate protein expression and many cellular, biochemical and physiological processes. According to their size ncRNAs are classified into small ncRNAs (<200 nucleotides long), including microRNAs (miRNAs) and long ncRNAs (lncRNAs), with a length >200 base pair, and circular RNAs (circRNA) (Hombach and Kretz 2016; St. Laurent et al. 2015). P-element-induced wimpy testis (PIWI-interacting RNAs (piRNAs)), a small ncRNAs once thought to be mainly functioning in germlines (Ozata et al. 2019), is now known to play an essential role in various diseases, including cancer, neurodegenerative diseases, and ageing (Huang and Wong 2021; Su et al. 2020; Wang et al. 2021a). In fact, the piRNA DQ541777 recruits DNMT3a to the CpG islands of the cdk5rap1 promoter, resulting in the methylation of cdk5rap1 and in the regulation of neuropathic pain in the spinal cord after CCI-induced NP. The regulatory effect of piRNA DQ541777 is suggested to be a novel element for future understanding and developing treatments against NP (Zhang et al. 2019). On the other hand, the aberrant expression in neural tissue of piRNAs (whether a disorder cause or consequence) remains a critical issue and its resolution might also facilitate applications for diagnostics and/or therapies in the future (Huang and Wong 2021). Indeed, a dysregulation of ncRNAs has been associated with a variety of neurodegenerative diseases, as well as NP after SCI, where gene-controlled processes can be stimulated or inhibited by ncRNAs (Li et al. 2019; Sámano et al. 2021; Zheng et al. 2022). However, the mechanisms through which those nc-RNAs contribute to the persistence of NP are not fully understood.

In a study using peripheral blood samples of SCI patients with or without NP analysis of lncRNA and miRNA expression levels, revealed a total of 289 lncRNAs and 18197 miRNAs. The co-expression network was investigated by KEGG (Kyoto Encyclopaedia of Genes and Genomes) analysis, where three KEGG pathways, seven genes directly and two lncRNAs were found to be involved in the NP pathway (E2F1, MAX, MITF, CTNNA1, ADORA2B, GRIK3, OXTR, LINC01119, and LINC02447). This suggest that these genes and lncRNAs may play an important role in NP, and might serve as biomarkers of SCI-induced NP (Zhou et al. 2017).

Several ncRNAs have been shown to play a role in the pathogenesis of NP, since they have been identified in pain-related regions (e.g. DRG and spinal cord) of mouse, rat and human nervous system, although the specific effects of ncRNAs in NP remain largely unknown (Jiang et al. 2015; Pan et al. 2017; Ray et al. 2018; Song et al. 2020). Moreover, several reports demonstrated an aberrant expression of ncRNAs (up or downregulated) and the human homologues functions point to them being potential biomarkers for the diagnosis with a crucial role in the pathophysiological process of NP (Kalpachidou et al. 2020; Song et al. 2020).

All these data represented an innovative data integration analysis of a large number of small molecules that microarray technology can disclose with high impact on understanding NP pathogenesis. We are aware that these results are limited to bulk measurements, and recent reports pointed out the need to capture diverse responses of highly heterogeneous samples and even single-cell gene expression profiling for multiple screening at single-cell level (Shin et al. 2019) This approach should be important to decipher the pathway mechanisms of the spinal dorsal horn that are complex and have implications for the pathological enhancement of NP and are also implicated in cell interactions and cell plasticity during pain development (Mauceri 2022). Increasing evidence sheds light on the interactions among ncRNAs and mRNAs in regulating the cellular functions in neurons and glia during NP development (Jin et al. 2018; Song et al. 2020; Xia et al. 2018). Additionally, on the basis of the functional properties of differentially expressed ncRNAs, special focus has been made on better understanding the role and the interactions with a type of lncRNAs, termed circRNA, especially during the NP development (Kalpachidou et al. 2020; Song et al. 2020; Xu et al. 2021b; Zheng et al. 2022).

CircRNAs are expressed from known protein-coding genes (Guo et al. 2014), and at present, four types have been identified: intergenic circRNAs, exo-intron circRNAs (EIciRNAs), and circRNAs from introns and exonic circRNAs (ecircRNAs) (Eger et al. 2018; Zhang et al. 2018).

While pharmacological, nonpharmacological and interventional therapies have been currently used for the management of chronic pain, treatments remain inadequate for a number of patients. In this regard, application of ncRNAs medicine based on miRNAs and lncRNAs to pain prevention or relief represents a new strategy (Jiang et al. 2015; Liu et al. 2017; Roganović and Petrović 2022). Several reports have shown that ncRNAs are involved in inflammatory signaling (Li et al. 2021b), and have indicated new avenues for ncRNAs in diagnostics and therapeutic interventions (Bali and Kuner 2014). Particularly for treating NP, small-molecule targeted therapies using miRNAs or lncRNAs have been explored to edit ncRNA genes with effects that could include disrupting regulatory, structural or functional motifs. Another therapeutic strategy has been targeted to regulate the protein-coding genes at epigenetic, transcriptional or post-transcriptional level (Qureshi and Mehler 2013; Tang et al. 2019). Although some RNA based-therapies have already been approved by the Food Drug Administration (FDA) or the European Medicines Agency (EMA) to treat central or peripheral nervous system diseases (Damase et al. 2021; Winkle et al. 2021), their use in pain research remains limited. This is because dysregulation of ncRNAs has been reported in various regions of different animal models (Bali and Kuner 2014; Li et al. 2021b). Additionally, there may be differences between in vitro and in vivo models and the translational potential is still unknown in humans (Song et al. 2020; Winkle et al. 2021; Wu et al. 2019). Within this framework further work on ncRNAs is needed to clarify their clinical application, efficiency, risks and tolerability. lncRNAs that are the computational construction of interaction between lncRNAs-circRNAs–miRNAs–mRNAs can provide new directions and potential therapeutic targets (Song et al. 2020; Winkle et al. 2021).

To date, circRNAs have been demonstrated to be neuronal specific and to have a close dynamic temporal regulation related to cell death mechanism upon injury (Hanan et al. 2017; Kalpachidou et al. 2020; Ohnishi et al. 2022; Rybak-Wolf et al. 2015). circRNAs are abundant lncRNAs and are well conserved: they consist of a closed continuous loop, which leads to high stability. Most of the circRNAs have been reported to regulate target gene transcription, such as the miRNA expression during the pathological processes in NP (Guo et al. 2020). It is important to emphasize that discrepancies between in vitro and in vivo experiments have arisen. In a recent review, Xu et al. point out that the differences among the differential expression of lncRNAs in many investigations could be due to the lack of specificity of lncRNAs in different NP animal models, and to the source of cells used for each study; these could also be limitations for the development of clinical trials using ncRNAs (Xu et al. 2021b; Zhou et al. 2017). Besides it is still unknown whether hypersensitivity may depend on sexual dimorphism (Xu et al. 2021b).

In recent years, circRNAs have been shown to crosstalk with miRNAs in a negative way, by sponging (Hansen et al. 2013; Panda 2018; Zhang et al. 2018), or in a positive manner, by direct sequence interactions (Piwecka et al. 2017). Thus, Zhou and collaborators have described that the interaction among some lncRNAs, miRNAs, circRNAs, and mRNAs plays an important role during NP pathogenesis in a model of spared nerve injury (SNI). The sequencing and bioinformatics analysis of the expression patterns of ncRNAs has revealed that a total of 134 lncRNAs (12 miRNAs, 188 circRNAs and 1066 mRNAs) is significantly regulated in the spinal cord 14 days after SNI-induced NP. The functional prediction suggests that the regulatory network of lncRNA-miRNA-mRNA and circRNA-miRNA-mRNA are involved during NP pathogenesis, via signalling pathways such as PI3K-Akt, focal adhesion, ECM-receptor interaction, protein digestion and absorption (Zhou et al. 2017). With these data, many investigators have proposed that lncRNAs, including circRNAs are dysregulated during the pathogenesis of NP and several novel points of regulation have been proposed. Particularly, circRNAs due to their biological functions are likely to be used as diagnostic biomarkers in NP or to be considered as novel potential therapeutic targets (Zheng et al. 2022; Zhou et al. 2017).

Cellular and molecular mechanisms regulated by circRNAs to manage pain

In 2017 Cao and colleagues have identified the transcriptome of circRNAs in spinal dorsal horn of a model of CCI neuropathic pain in rats. By circRNAs microarrays they have detected a total of 12,770 circRNAs, from which 469 circRNAs are differentially expressed in CCI rats. These authors have reported 363 circRNAs significantly upregulated and 106 downregulated. Specifically, three circRNAs: rno_circRNA_013779, rno_circRNA_013779, and rno_circRNA_013779, overexpressed over 10 fold in the CCI group. Later, gene ontology (GO) and KEGG pathway analyses have revealed several predicted target genes involved in biological mechanisms, cellular and molecular mechanisms associated with Hippo signaling pathways. Additionally, the circRNA–microRNA–mRNA network analysis has shown the circRNA_008008 and circRNA_013779 as the two largest nodes in the circRNA–microRNA interaction mode. These observations indicate that after CCI insult there are several changes in circRNAs expression in the rat spinal dorsal horn (Cao et al. 2017; Zheng et al. 2022).

Another bioinformatic study revealed that the expression profiling of circRNAs in NP has been associated with several molecular pathways such as PI3K/Akt/mTOR, Wnt/β-catenin, and MAPK signaling by complex sequence of events that are tightly regulated (Salami et al. 2022). In this context the review published by Yash and colleagues has described the landscape of miRNA regulatory network (interactome) and their signal transduction pathways targets, such as IRAK/TRAF6, TLR4/NF-kB, MAPK, TGFb, and TLR5 in NP, which have been proposed as potential biomarkers for diagnosis, prognosis and therapeutic intervention in NP (Gada et al. 2022).

The sponge function of circRNAs by differentially interacting with other miRNAs has been proposed to play a pivotal role during development of NP (Figure 1). For its part, ciRS-7 expression is found in the rat spinal cord dorsal horn of the CCI model of NP. The knockdown of ciRS-7 attenuates microglia activation and expression of pro-inflammatory cytokines, while ciRS-7 targeting miR-135a-5p is associated with NP progress via autophagy. The data suggest that targeting either ciRS-7 or miR-135a-5p could be effective in alleviating NP through suppressing neuroinflammation (Cai et al. 2020). Another circRNA found to be upregulated in the dorsal spinal cord after CCI model in rats, is the circ_0005075: after its knockdown NP behaviours (thermal hyperalgesia and mechanical allodynia) are suppressed and neuroinflammation inhibited through targeting of TNF-α, IL-6, IL-10, and cyclooxygenase (COX)-2. This highlights that the sponging function of cmiR-151a-3p by circ_0005075 depresses NOTCH2 to mediate the neuroinflammation and NP development (Zhang et al. 2021).

Figure 1: 
A schematic drawing of circRNA biogenesis of several miRNAs against degeneration and development upon SCI. Several circRNA can act as a compensatory mechanism to achieve NP relief by sponge function regulation. Created with Biorender.com.
Figure 1:

A schematic drawing of circRNA biogenesis of several miRNAs against degeneration and development upon SCI. Several circRNA can act as a compensatory mechanism to achieve NP relief by sponge function regulation. Created with Biorender.com.

The effect of circRNAs on microglia and astrocytes following SCI has been documented (Chen et al. 2021), particularly the expression of circPrkcsh shown to be upregulated in astrocytes of mouse injured spinal cord. The circPrkcsh knockout decreases inflammatory response after SCI through the chemokine CC motif ligand 2 (CCL2) expression via miR-488 upregulation (Chen et al. 2022a). In a SCI mouse model the circ-Usp10/miR-152-5p/CD84 axis activates microglial cells, while silencing circ-Usp10 significantly reduces neuronal death of cell line HT22. Thus, circ- Usp10 may promote microglial activation and induce neuronal death by targeting miR-152-5p/CD84 (Tong et al. 2021). The role of circRNA in NP remains poorly understood with a report showing that circSMEK1 facilitates NP inflammation and microglia polarization by modulating miR-216a-5p/TXNIP axis in the NP rat and cell models (Xin et al. 2021). Understanding the glia–glia interactions with sensory neurons together with circRNAs regulation could contribute to identifying the molecular mechanisms of pain signaling, and may provide future therapeutic options to target pain.

Importantly, the circular RNAs expression of both cZRANB1 and circZNF609 is decreased in CCI model in rats. In the study by Wei et al. the upregulation of cZRANB1 alleviates neuropathic pain via modulating Wnt5a/β-Catenin signalling levels by sponging miR-24-3p/LPAR3 axis (Wei et al. 2020). For their part, Li and colleagues showed that circZNF609 promotes inflammatory factors expression, aggravating NP progression via miR-22-3p/ENO1 axis in CCI rat models further confirmed because the functional knockdown of circZNF609 alleviates thermal hyperalgesia and mechanical allodynia through IL6, TNF-α and IL-1 (Li et al. 2020).

On the other hand, in a recent study using microarray analysis and cell cultures from 45 idiopathic scoliosis patients, 25 females and 20 males, has shown that circRNA-CIDN is down-regulated under compression stress. Indeed, the downregulation of extracellular matrix and apoptosis is achieved by sequestering with miR-34a-5p via repressing the silent mating type information regulation 2 homolog 1 (SIRT1) (Xiang et al. 2020). Further evidence for circRNAs role in NP has been obtained by analysing the role of cytoplasmic circAnks1a that acts as a miRNA sponge in miR-324-3p and mediates posttranscriptional regulation of VEGFB expression after sciatic nerve ligation (Zhang et al. 2019). In addition, growing evidence has also shown that miRNAs and circRNAs regulate the occurrence and progression of the intervertebral disc degeneration (IDD) triggered by low back pain (LBP). In a very elegant study Li and collaborators have identified the upregulation of circ-FAM169A in cytoplasm of degenerative nucleus pulposus cells (NPC), which is negatively correlated with the expression of miR-583. Besides that, the prediction analysis has shown that the construction circ-FAM169A-miR-583-mRNAs network is related to the extracellular matrix metabolism and apoptosis, where miR-583 negatively regulates Sox9 mRNA and other proteins in NPCs. With these data, Li et al. suggests that the function of circ-FAM169A as ceRNA is to sponge miR-583, which in turn will modulate the pathogenesis of IDD. Although circ-FAM169A-miR-583-mRNAs was identified as key network involved in IDD, the significant role of the circ-FAM169A-miR-583 requires further investigation (Li et al. 2021a). Recent evidence obtained from 15 patients suffering from IDD and low back pain, has investigated the expression and sponge function of circRNAs in spinal tissues in vivo and in vitro. Specifically, the biological function and differential expression of circRNA are upregulated (circ-4099, circRNA_104670, circ-FAM169A, and circ-TIMP2) and/or downregulated (circSEMA4B, circ-GRB10, circVMA21, and circRNA-CIDN) suggesting a key role in several processes such as apoptosis, proliferation, and senescence by acting as compensatory mechanism against degeneration and pain development. Indeed, more evidence obtained from patients suffering from IDD and low back pain, points to the biological function of ccirc_0040039 and circ_0004354 in modulating inflammatory response, growth inhibition, and extracellular matrix components and, therefore, a crucial role in disc degeneration. These data have demonstrated, by bioinformatics analysis of microarray dataset, that the mechanism of these circRNAs is to induce mixed apoptosis and inflammatory response due to a competitive adsorption of miR-345-3p (Li et al. 2022). Finally, the gene expression profile of circRNA in certain regions of post-mortem brain tissue of 42 Parkinson’s disease patients was recently analysed (Wu and Kao 2016). This report shows the modulation of several pathways such as neurodegeneration and synaptic transmission, but probably by cooperating with RNA processing of gene products to exacerbate or limit neurodegeneration. Indeed, the cooperation of different cells types, that are known to modulate neuronal functions, may exacerbate damage under diverse insults (Wu and Kao 2016). In summary, this report shows that circRNA probably operates not only by sponging miRNAs but also contributes to complex events involved in altered checkpoints (circularized splicing, miRNA regulation, and protein decline) which may amplify the functional impact alongside the stoichiometry of those molecules (Hanan et al. 2020). circRNAs are increasingly recognized as important regulatory molecules of various cellular and molecular processes (Xu et al. 2021a). Dysregulated functions have been reported during pain development and in response to SCI (Jiang et al. 2022). Because circRNAs are highly stable and usually expressed in a tissue, cell and disease type-specific manner, they have been currently proposed and explored as potential therapeutic targets (Ma et al. 2020). In this context, gain-or-loss of function approaches are performed using circRNAs expression plasmids or RNA-interference-based strategies. In particular, spinal injection of circAnks1a siRNA has been found to alleviate pain-like behavior in rats (Zhang et al. 2019). circHIPK3 shRNA can reduce NP in diabetic rats (Wang et al. 2018), and the circRNA.2837 inhibitor decreases pain in the sciatic nerve injured rat model by sponging miR-34 and inducing autophagy (Zhou et al. 2018). circRNAs have unique expression signatures and play crucial roles in transcriptional and posttranscriptional mechanisms, leading to the development of circRNA-based therapeutics.

Conclusions

NP as a consequence of SCI is a condition with currently limited effective treatment. Unfortunately, patients describe NP as an unbearable condition with long-term effects that affect their quality of life. At present, the research on NP related to SCI is mostly concentrated in rodent animal models, and recent studies have provided data in which cellular and molecular mechanisms are induced by many dysregulated gene expressions, with the lncRNAs as the regulatory factors proposed to play a relevant role during the pathomechanisms involved in NP. Particularly, growing evidence has emerged demonstrating dysregulation of several circRNAs involved in NP development (Figure 1). circRNAs have gained pathophysiological relevance, because of their synthesis by a noncanonical splicing event, their stability for up to 48 h in cells, and their specificity function as sponge of miRNAs. In this paper, we summarize recent data concerning circRNAs modulation by sponging miRNAs and suppressing or promoting cellular and molecular pathways and the interaction with mRNAs and proteins (Figure 2). Indeed, up to today, management of NP has been concentrated to reduce the toxicity and addictive potential; hence, circRNAs regulation may unveil certain molecular mechanisms involved in NP (hyperalgesia, allodynia and spontaneous pain) and can be used as novel potential effectors for chronic pain treatment. Nevertheless, in this review we have highlighted recent data supporting the idea of circRNAs acting as reliable biomarkers to manage NP after SCI. Therefore, translational studies using expression profile analysis of circRNAs, probably from body fluids, point out the importance for novel pharmacological treatments with high therapeutic value. Altogether, these data demonstrate that circRNAs are likely to be used as potential diagnostic in NP or to be considered as novel potential therapeutic targets after spinal injuries. Finally, all of this knowledge supports the proposal that circRNAs could be good therapeutic options for biomarkers and/or novel targets to NP relief and treatment, since unlike other treatments, they do not show toxicity or potential addiction.

Figure 2: 
A schematic diagram of circRNA sponge functional regulation due to several proteins interactions and the main cellular processes involved in NP modulation, such as apoptosis, proliferation, and senescence. Created with Biorender.com.
Figure 2:

A schematic diagram of circRNA sponge functional regulation due to several proteins interactions and the main cellular processes involved in NP modulation, such as apoptosis, proliferation, and senescence. Created with Biorender.com.


Corresponding authors: Graciela L. Mazzone, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Av. Pte. Perón 1500, B1629AHJ, Pilar, Buenos Aires, Argentina, E-mail: ; and Cynthia Sámano, Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa. Avenida Vasco de Quiroga 4871, Col. Santa Fe Cuajimalpa. Alcaldía Cuajimalpa de Morelos, C.P. 05348, Ciudad de México, México, E-mail:

Funding source: Ministry of Science, Technology and Productive Innovation of Argentina through the Fund for Scientific and Technological Research

Award Identifier / Grant number: PICT-2020-SERIEA-00928

Award Identifier / Grant number: PICT-2020-SERIEA-00030

Funding source: Departamento de Ciencias Naturales (DCN), de la División de Ciencias Naturales e Ingeniería (DCNI) de la UAM-Cuajimalpa

Funding source: Croatian Science Foundation grant

Award Identifier / Grant number: IP-2016-06-7060

Funding source: Universidad Austral, CONICET

Funding source: UniRi research

Award Identifier / Grant number: prirod-sp-20-38-2687

Award Identifier / Grant number: uniri-biomed-18-258-1427

Funding source: IBRO Collaborative Research Grant

Acknowledgment

The authors would like to thank Prof Andrea Nistri and Dr. Carly McCarthy for the helpful comments on the manuscript.

  1. Author contributions: GM and CS contributed to the conception of principal ideas, wrote the first draft of the manuscript and performed the graphic design. MM and MFC wrote some sections of the manuscript, and thoroughly proofread the last version. All authors contributed to the article and approved the submitted version.

  2. Research funding: GM was supported by Universidad Austral, CONICET, Ministry of Science, Technology and Productive Innovation of Argentina through the Fund for Scientific and Technological Research (FONCYT, PICT-2020-SERIEA-00928) and IBRO Collaborative Research Grant. MFC was supported by Universidad Austral, CONICET and the Ministry of Science Argentina (PICT-2020-SERIEA-00030). MM was supported by Croatian Science Foundation grant IP-2016-06-7060 and UniRi research support uniri-biomed-18-258-1427 and prirod-sp-20-38-2687. CS was supported by the Departamento de Ciencias Naturales (DCN) de la División de Ciencias Naturales e Ingeniería (DCNI) de la UAM-Cuajimalpa (Divisional Project 47301025).

  3. Conflict of interest statement: The authors declare no conflict of interest.

References

Ahuja, C.S., Wilson, J.R., Nori, S., Kotter, M.R.N., Druschel, C., Curt, A., and Fehlings, M.G. (2017). Traumatic spinal cord injury. Nat. Rev. Dis. Prim. 3: 17018, https://doi.org/10.1038/nrdp.2017.18.Search in Google Scholar PubMed

Bali, K.K. and Kuner, R. (2014). Noncoding RNAs: key molecules in understanding and treating pain. Trends Mol. Med. 20: 437–448, https://doi.org/10.1016/j.molmed.2014.05.006.Search in Google Scholar PubMed PubMed Central

Berg, M.V.D., Castellote, J.M., Mahillo-Fernandez, I., and de Pedro-Cuesta, J. (2011). Incidence of traumatic spinal cord injury in Aragón, Spain (1972–2008). J. Neurotrauma 28: 469–477, https://doi.org/10.1089/neu.2010.1608.Search in Google Scholar PubMed

Berta, T., Qadri, Y.J., Chen, G., and Ji, R.R. (2016). Microglial signaling in chronic pain with a special focus on caspase 6, p38 MAP kinase, and sex dependence. J. Dent. Res. 95: 1124–1131, https://doi.org/10.1177/0022034516653604.Search in Google Scholar PubMed PubMed Central

Bianchi, M.S., Calvo, V., Chasseing, N.A., Lago, N., Libertun, C., Montaner, A.D., and Lux-Lantos, V.A. (2012). Oligodeoxynucleotide IMT504: lack of effect on immune parameters during islet regeneration in single dose streptozotocin-induced diabetes: IMT504, STZ-induced diabetes: IDO, TSG-6. Diabetes Metab. Res. Rev. 28: 156–163, https://doi.org/10.1002/dmrr.1296.Search in Google Scholar PubMed

Boroujerdi, A., Zeng, J., Sharp, K., Kim, D., Steward, O., and Luo, D.Z. (2011). Calcium channel alpha-2-delta-1 protein upregulation in dorsal spinal cord mediates spinal cord injury-induced neuropathic pain states. Pain 152: 649–655, https://doi.org/10.1016/j.pain.2010.12.014.Search in Google Scholar PubMed PubMed Central

Britch, S.C. and Walsh, S.L. (2022). Treatment of opioid overdose: current approaches and recent advances. Psychopharmacology 239: 2063–2081, https://doi.org/10.1007/s00213-022-06125-5.Search in Google Scholar PubMed PubMed Central

Brockie, S., Hong, J., and Fehlings, M.G. (2021). The Role of microglia in modulating neuroinflammation after spinal cord injury. Int. J. Mol. Sci. 22: 9706, https://doi.org/10.3390/ijms22189706.Search in Google Scholar PubMed PubMed Central

Bryce, T.N., Biering-Sørensen, F., Finnerup, N.B., Cardenas, D.D., Defrin, R., Ivan, E., Lundeberg, T., Norrbrink, C., Richards, J.S., Siddall, P., et al.. (2012a). International spinal cord injury pain (ISCIP) classification: Part 2. Initial validation using vignettes. Spinal Cord 50: 404–412, https://doi.org/10.1038/sc.2012.2.Search in Google Scholar PubMed

Bryce, T.N., Biering-Sørensen, F., Finnerup, N.B., Cardenas, D.D., Defrin, R., Lundeberg, T., Norrbrink, C., Richards, J.S., Siddall, P., Stripling, T., et al.. (2012b). International spinal cord injury pain classification: part I. Background and description. Spinal Cord 50: 413–417, https://doi.org/10.1038/sc.2011.156.Search in Google Scholar PubMed

Burke, D., Fullen, B.M., Stokes, D., and Lennon, O. (2017). Neuropathic pain prevalence following spinal cord injury: a systematic review and meta-analysis. Eur. J. Pain 21: 29–44, https://doi.org/10.1002/ejp.905.Search in Google Scholar PubMed

Burnstock, G. (2020). Introduction to purinergic signalling in the brain. In: Barańska, J. (Ed.), Glioma signaling. Springer International Publishing, Cham, pp. 1–12.10.1007/978-3-030-30651-9_1Search in Google Scholar PubMed

Cai, W., Zhang, Y., and Su, Z. (2020). ciRS-7 targeting miR-135a-5p promotes neuropathic pain in CCI rats via inflammation and autophagy. Gene 736: 144386, https://doi.org/10.1016/j.gene.2020.144386.Search in Google Scholar PubMed

Cao, S., Deng, W., Li, Y., Qin, B., Zhang, L., Yu, S., Xie, P., Xiao, Z., and Yu, T. (2017). Chronic constriction injury of sciatic nerve changes circular RNA expression in rat spinal dorsal horn. J. Pain Res. 10: 1687–1696, https://doi.org/10.2147/JPR.S139592.Search in Google Scholar PubMed PubMed Central

Casadei, M., Fiore, E., Rubione, J., Domínguez, L.M., Coronel, M.F., Leiguarda, C., García, M., Mazzolini, G., Villar, M.J., Montaner, A., et al.. (2022). IMT504 blocks allodynia in rats with spared nerve injury by promoting the migration of mesenchymal stem cells and by favoring an anti-inflammatory milieu at the injured nerve. Pain 163: 1114–1129, https://doi.org/10.1097/j.pain.0000000000002476.Search in Google Scholar PubMed PubMed Central

Chen, G., Luo, X., Qadri, M.Y., Berta, T., and Ji, R.-R. (2018). Sex-dependent glial signaling in pathological pain: distinct roles of spinal microglia and astrocytes. Neurosci. Bull. 34: 98–108, https://doi.org/10.1007/s12264-017-0145-y.Search in Google Scholar PubMed PubMed Central

Chen, J.-N., Zhang, Y.-N., Tian, L.-G., Zhang, Y., Li, X.-Y., and Ning, B. (2022a). Down-regulating circular RNA Prkcsh suppresses the inflammatory response after spinal cord injury. Neural Regen. Res. 17: 144–151, https://doi.org/10.4103/1673-5374.314114.Search in Google Scholar PubMed PubMed Central

Chen, M., Lai, X., Wang, X., Ying, J., Zhang, L., Zhou, B., Liu, X., Zhang, J., Wei, G., and Hua, F. (2021). Long non-coding RNAs and circular RNAs: insights into microglia and astrocyte mediated neurological diseases. Front. Mol. Neurosci. 14: 745066, https://doi.org/10.3389/fnmol.2021.745066.Search in Google Scholar PubMed PubMed Central

Chen, Y., Zhou, Y., Li, X.-C., Ma, X., Mi, W.-L., Chu, Y.-X., Wang, Y.-Q., and Mao-Ying, Q.-L. (2022b). Neuronal GRK2 regulates microglial activation and contributes to electroacupuncture analgesia on inflammatory pain in mice. Biol. Res. 55: 5, https://doi.org/10.1186/s40659-022-00374-6.Search in Google Scholar PubMed PubMed Central

Chen, Y.-W., Li, Y.-T., Chen, Y.C., Li, Z.-Y., and Hung, C.-H. (2012). Exercise training attenuates neuropathic pain and cytokine expression after chronic constriction injury of rat sciatic nerve. Anesth. Analg. 114: 1330–1337, https://doi.org/10.1213/ANE.0b013e31824c4ed4.Search in Google Scholar PubMed

Collins, S., Sigtermans, M.J., Dahan, A., Zuurmond, W.W.A., and Perez, R.S.G.M. (2010). NMDA receptor antagonists for the treatment of neuropathic pain. Pain Med. 11: 1726–1742, https://doi.org/10.1111/j.1526-4637.2010.00981.x.Search in Google Scholar PubMed

Colloca, L., Ludman, T., Bouhassira, D., Baron, R., Dickenson, A.H., Yarnitsky, D., Freeman, R., Truini, A., Attal, N., Finnerup, N.B., et al.. (2017). Neuropathic pain. Nat. Rev. Dis. Prim. 3: 17002, https://doi.org/10.1038/nrdp.2017.2.Search in Google Scholar PubMed PubMed Central

Coronel, M.F., Hernando-Insúa, A., Rodriguez, J.M., Elias, F., Chasseing, N.A., Montaner, A.D., and Villar, M.J. (2008). Oligonucleotide IMT504 reduces neuropathic pain after peripheral nerve injury. Neurosci. Lett. 444: 69–73, https://doi.org/10.1016/j.neulet.2008.07.045.Search in Google Scholar PubMed

Crown, E.D. (2012). The role of mitogen activated protein kinase signaling in microglia and neurons in the initiation and maintenance of chronic pain. Exp. Neurol. 234: 330–339, https://doi.org/10.1016/j.expneurol.2011.10.019.Search in Google Scholar PubMed

Damase, T.R., Sukhovershin, R., Boada, C., Taraballi, F., Pettigrew, R.I., and Cooke, J.P. (2021). The limitless future of RNA therapeutics. Front. Bioeng. Biotechnol. 9: 628137, https://doi.org/10.3389/fbioe.2021.628137.Search in Google Scholar PubMed PubMed Central

Detloff, M.R., Smith, E.J., Quiros Molina, D., Ganzer, P.D., and Houlé, J.D. (2014). Acute exercise prevents the development of neuropathic pain and the sprouting of non-peptidergic (GDNF- and artemin-responsive) c-fibers after spinal cord injury. Exp. Neurol. 255: 38–48, https://doi.org/10.1016/j.expneurol.2014.02.013.Search in Google Scholar PubMed PubMed Central

Donnelly, C.R., Andriessen, A.S., Chen, G., Wang, K., Jiang, C., Maixner, W., and Ji, R.-R. (2020). Central nervous system targets: glial cell mechanisms in chronic pain. Neurotherapeutics 17: 846–860, https://doi.org/10.1007/s13311-020-00905-7.Search in Google Scholar PubMed PubMed Central

Dubový, P., Klusáková, I., Svíženská, I., and Brázda, V. (2010). Spatio-temporal changes of SDF1 and its CXCR4 receptor in the dorsal root ganglia following unilateral sciatic nerve injury as a model of neuropathic pain. Histochem. Cell Biol. 133: 323–337, https://doi.org/10.1007/s00418-010-0675-0.Search in Google Scholar PubMed

Eger, N., Schoppe, L., Schuster, S., Laufs, U., and Boeckel, J.-N. (2018). Circular RNA splicing. In: Xiao, J. (Ed.), Circular RNAs. Springer Singapore, Singapore, pp. 41–52.10.1007/978-981-13-1426-1_4Search in Google Scholar PubMed

Elias, F., Flo, J., Lopez, R.A., Zorzopulos, J., Montaner, A., and Rodriguez, J.m. (2003). Strong cytosine-guanosine-independent immunostimulation in humans and other primates by synthetic oligodeoxynucleotides with PyNTTTTGT motifs. J. Immunol. 171: 3697–3704, https://doi.org/10.4049/jimmunol.171.7.3697.Search in Google Scholar PubMed

Erbacher, C., Vaknine, S., Moshitzky, G., Lobentanzer, S., Eisenberg, L., Evdokimov, D., Sommer, C., Greenberg, D.S., Soreq, H., and Üçeyler, N. (2022). Distinct CholinomiR blood cell signature as a potential modulator of the cholinergic system in women with fibromyalgia syndrome. Cells 11: 1276, https://doi.org/10.3390/cells11081276.Search in Google Scholar PubMed PubMed Central

Felix, R., Muñoz-Herrera, D., Corzo-López, A., Fernández-Gallardo, M., Leyva-Leyva, M., González-Ramírez, R., and Sandoval, A. (2022). Ion channel long non-coding RNAs in neuropathic pain. Pflugers Arch - Eur. J. Physiol. 474: 457–468, https://doi.org/10.1007/s00424-022-02675-x.Search in Google Scholar PubMed

Finnerup, N.B. (2013). Pain in patients with spinal cord injury. Pain 154: S71–S76, https://doi.org/10.1016/j.pain.2012.12.007.Search in Google Scholar PubMed

Finnerup, N., Johannesen, I., Sindrup, S., Bach, F., and Jensen, T. (2001). Pain and dysesthesia in patients with spinal cord injury: a postal survey. Spinal Cord 39: 256–262, https://doi.org/10.1038/sj.sc.3101161.Search in Google Scholar PubMed

Finnerup, N.B., Kuner, R., and Jensen, T.S. (2021). Neuropathic pain: from mechanisms to treatment. Physiol. Rev. 101: 259–301, https://doi.org/10.1152/physrev.00045.2019.Search in Google Scholar PubMed

Gada, Y., Pandey, A., Jadhav, N., Ajgaonkar, S., Mehta, D., and Nair, S. (2022). New vistas in microRNA regulatory interactome in neuropathic pain. Front. Pharmacol. 12: 778014, https://doi.org/10.3389/fphar.2021.778014.Search in Google Scholar PubMed PubMed Central

Geng, S.-J., Liao, F.-F., Dang, W.-H., Ding, X., Liu, X.-D., Cai, J., Han, J.-S., Wan, Y., and Xing, G.-G. (2010). 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. Exp. Neurol. 222: 256–266, https://doi.org/10.1016/j.expneurol.2010.01.003.Search in Google Scholar PubMed

Goudet, C., Chapuy, E., Alloui, A., Acher, F., Pin, J.-P., and Eschalier, A. (2008). Group III metabotropic glutamate receptors inhibit hyperalgesia in animal models of inflammation and neuropathic pain. Pain 137: 112–124, https://doi.org/10.1016/j.pain.2007.08.020.Search in Google Scholar PubMed

Guo, H.-Y., Guo, M.-K., Wan, Z.-Y., Song, F., and Wang, H.-Q. (2020). Emerging evidence on noncoding-RNA regulatory machinery in intervertebral disc degeneration: a narrative review. Arthritis Res. Ther. 22: 270, https://doi.org/10.1186/s13075-020-02353-2.Search in Google Scholar PubMed PubMed Central

Guo, J.U., Agarwal, V., Guo, H., and Bartel, D.P. (2014). Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 15: 409, https://doi.org/10.1186/s13059-014-0409-z.Search in Google Scholar PubMed PubMed Central

Hanan, M., Soreq, H., and Kadener, S. (2017). CircRNAs in the brain. RNA Biol. 14: 1028–1034, https://doi.org/10.1080/15476286.2016.1255398.Search in Google Scholar PubMed PubMed Central

Hanan, M., Simchovitz, A., Yayon, N., Vaknine, S., Cohen-Fultheim, R., Karmon, M., Madrer, N., Rohrlich, T.M., Maman, M., Bennett, E.R., et al.. (2020). A Parkinson’s disease CircRNAs resource reveals a link between circSLC8A1 and oxidative stress. EMBO Mol. Med. 12: e11942, https://doi.org/10.15252/emmm.201911942.Search in Google Scholar PubMed PubMed Central

Hansen, T.B., Jensen, T.I., Clausen, B.H., Bramsen, J.B., Finsen, B., Damgaard, C.K., and Kjems, J. (2013). Natural RNA circles function as efficient microRNA sponges. Nature 495: 384–388, https://doi.org/10.1038/nature11993.Search in Google Scholar PubMed

Hasegawa, M., Fujimoto, Y., Lucas, P.C., Nakano, H., Fukase, K., Núñez, G., and Inohara, N. (2008). A critical role of RICK/RIP2 polyubiquitination in Nod-induced NF-κB activation. EMBO J. 27: 373–383, https://doi.org/10.1038/sj.emboj.7601962.Search in Google Scholar PubMed PubMed Central

Hassanpour, K., Hotz-Boendermaker, S., Dokladal, P., and Curt, A. (2012). Low depressive symptoms in acute spinal cord injury compared to other neurological disorders. J. Neurol. 259: 1142–1150, https://doi.org/10.1007/s00415-011-6316-2.Search in Google Scholar PubMed

Hewitt, D.J. (2000). The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin. J. Pain 16: S73–S79, https://doi.org/10.1097/00002508-200006001-00013.Search in Google Scholar PubMed

Hombach, S. and Kretz, M. (2016). Non-coding RNAs: classification, biology and functioning. In: Slaby, O. and Calin, G.A. (Eds.), Non-coding RNAs in colorectal cancer. Springer International Publishing, Cham, pp. 3–17.10.1007/978-3-319-42059-2_1Search in Google Scholar PubMed

Huang, X. and Wong, G. (2021). An old weapon with a new function: PIWI-interacting RNAs in neurodegenerative diseases. Transl. Neurodegener. 10: 9, https://doi.org/10.1186/s40035-021-00233-6.Search in Google Scholar PubMed PubMed Central

Hunt, C., Moman, R., Peterson, A., Wilson, R., Covington, S., Mustafa, R., Murad, M.H., and Hooten, W.M. (2021). Prevalence of chronic pain after spinal cord injury: a systematic review and meta-analysis. Reg. Anesth. Pain Med. 46: 328–336, https://doi.org/10.1136/rapm-2020-101960.Search in Google Scholar PubMed

Jiang, B.-C., Sun, W.-X., He, L.-N., Cao, D.-L., Zhang, Z.-J., and Gao, Y.-J. (2015). Identification of IncRNA expression profile in the spinal cord of mice following spinal nerve ligation-induced neuropathic pain. Mol. Pain 11: 43, https://doi.org/10.1186/s12990-015-0047-9.Search in Google Scholar PubMed PubMed Central

Jiang, M., Wang, Y., Wang, J., Feng, S., and Wang, X. (2022). The etiological roles of miRNAs, lncRNAs, and circRNAs in neuropathic pain: a narrative review. J. Clin. Lab. Anal. 36: e24592, https://doi.org/10.1002/jcla.24592.Search in Google Scholar PubMed PubMed Central

Jin, H., Du, X., Zhao, Y., and Xia, D. (2018). XIST/miR‐544 axis induces neuropathic pain by activating STAT3 in a rat model. J. Cell. Physiol. 233: 5847–5855, https://doi.org/10.1002/jcp.26376.Search in Google Scholar PubMed

Johansson, E., Luoto, T.M., Vainionpää, A., Kauppila, A.-M., Kallinen, M., Väärälä, E., and Koskinen, E. (2021). Epidemiology of traumatic spinal cord injury in Finland. Spinal Cord 59: 761–768, https://doi.org/10.1038/s41393-020-00575-4.Search in Google Scholar PubMed PubMed Central

Jongen, J.L.M., Hans, G., Benzon, H.T., Huygen, F., and Hartrick, C.T. (2014). Neuropathic pain and pharmacological treatment. Pain Pract. 14: 283–295, https://doi.org/10.1111/papr.12085.Search in Google Scholar PubMed

Kalpachidou, T., Kummer, K.K., and Kress, M. (2020). Non-coding RNAs in neuropathic pain. Neuronal Signal. 4: NS20190099, https://doi.org/10.1042/NS20190099.Search in Google Scholar PubMed PubMed Central

Kelly, M.J., Breathnach, C., Tracey, K.J., and Donnelly, S.C. (2022). Manipulation of the inflammatory reflex as a therapeutic strategy. Cell Rep. Med. 3: 100696, https://doi.org/10.1016/j.xcrm.2022.100696.Search in Google Scholar PubMed PubMed Central

Knerlich-Lukoschus, F., von der Ropp-Brenner, B., Lucius, R., Mehdorn, H.M., and Held-Feindt, J. (2011). Spatiotemporal CCR1, CCL3(MIP-1α), CXCR4, CXCL12(SDF-1α) expression patterns in a rat spinal cord injury model of posttraumatic neuropathic pain: laboratory investigation. J. Neurosurg. Spine 14: 583–597, https://doi.org/10.3171/2010.12.SPINE10480.Search in Google Scholar PubMed

Kobayashi, K., Yamanaka, H., Fukuoka, T., Dai, Y., Obata, K., and Noguchi, K. (2008). P2Y12 receptor upregulation in activated microglia is a gateway of p38 signaling and neuropathic pain. J. Neurosci. 28: 2892–2902, https://doi.org/10.1523/JNEUROSCI.5589-07.2008.Search in Google Scholar PubMed PubMed Central

Kuner, R. (2010). Central mechanisms of pathological pain. Nat. Med. 16: 1258–1266, https://doi.org/10.1038/nm.2231.Search in Google Scholar PubMed

Lacagnina, M.J., Watkins, L.R., and Grace, P.M. (2018). Toll-like receptors and their role in persistent pain. Pharmacol. Ther. 184: 145–158, https://doi.org/10.1016/j.pharmthera.2017.10.006.Search in Google Scholar PubMed PubMed Central

Latremoliere, A. and Woolf, C.J. (2009). Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J. Pain 10: 895–926, https://doi.org/10.1016/j.jpain.2009.06.012.Search in Google Scholar PubMed PubMed Central

Leiguarda, C., Coronel, M.F., Montaner, A.D., Villar, M.J., and Brumovsky, P.R. (2018). Long-lasting ameliorating effects of the oligodeoxynucleotide IMT504 on mechanical allodynia and hindpaw edema in rats with chronic hindpaw inflammation. Neurosci. Lett. 666: 17–23, https://doi.org/10.1016/j.neulet.2017.12.032.Search in Google Scholar PubMed

Leiguarda, C., Potilinski, C., Rubione, J., Tate, P., Villar, M.J., Montaner, A., Bisagno, V., Constandil, L., and Brumovsky, P.R. (2021a). IMT504 provides analgesia by modulating cell infiltrate and inflammatory milieu in a chronic pain model. J. Neuroimmune Pharmacol. 16: 651–666, https://doi.org/10.1007/s11481-020-09971-2.Search in Google Scholar PubMed

Leiguarda, C., Villarreal, A., Potilinski, C., Pelissier, T., Coronel, M.F., Bayo, J., Ramos, A.J., Montaner, A., Villar, M.J., Constandil, L., et al.. (2021b). Intrathecal administration of an anti‐nociceptive non-CpG oligodeoxynucleotide reduces glial activation and central sensitization. J. Neuroimmune Pharmacol. 16: 818–834, https://doi.org/10.1007/s11481-021-09983-6.Search in Google Scholar PubMed

Li, L., Luo, Y., Zhang, Y., Wei, M., Zhang, M., Liu, H., and Su, Z. (2020). CircZNF609 aggravates neuropathic pain via miR-22-3p/ENO1 axis in CCI rat models. Gene 763: 145069, https://doi.org/10.1016/j.gene.2020.145069.Search in Google Scholar PubMed

Li, T., Chen, X., Zhang, C., Zhang, Y., and Yao, W. (2019). An update on reactive astrocytes in chronic pain. J. Neuroinflammation 16: 140, https://doi.org/10.1186/s12974-019-1524-2.Search in Google Scholar PubMed PubMed Central

Li, Y., Pan, D., Liu, S., Xing, X., Zhou, H., Zhang, B., Zhang, D., Li, B., Li, G., Tao, B., et al.. (2021a). Identification of circ-FAM169A sponges miR-583 involved in the regulation of intervertebral disc degeneration. J. Orthop. Transl. 26: 121–131, https://doi.org/10.1016/j.jot.2020.07.007.Search in Google Scholar PubMed PubMed Central

Li, Y., Wu, X., Li, J., Du, L., Wang, X., Cao, J., Li, H., Huo, Z., Li, G., Pan, D., et al.. (2022). Circ_0004354 might compete with circ_0040039 to induce NPCs death and inflammatory response by targeting miR-345-3p-FAF1/TP73 axis in intervertebral disc degeneration. Oxid. Med. Cell. Longev. 2022: 1–21, https://doi.org/10.1155/2022/2776440.Search in Google Scholar PubMed PubMed Central

Li, Z., Li, X., Jian, W., Xue, Q., and Liu, Z. (2021b). Roles of long non-coding RNAs in the development of chronic pain. Front. Mol. Neurosci. 14: 760964, https://doi.org/10.3389/fnmol.2021.760964.Search in Google Scholar PubMed PubMed Central

Liang, Y., Qiu, Y., Du, J., Liu, J., Fang, J., Zhu, J., and Fang, J. (2016). Inhibition of Spinal microglia and astrocytes contributes to the anti-allodynic effect of electroacupuncture in neuropathic pain induced by spinal nerve ligation. Acupunct. Med. 34: 40–47, https://doi.org/10.1136/acupmed-2015-010773.Search in Google Scholar PubMed

Liu, T., Gao, Y.-J., and Ji, R.-R. (2012). Emerging role of toll-like receptors in the control of pain and itch. Neurosci. Bull. 28: 131–144, https://doi.org/10.1007/s12264-012-1219-5.Search in Google Scholar PubMed PubMed Central

Liu, Z., Liang, Y., Wang, H., Lu, Z., Chen, J., Huang, Q., Sheng, L., Ma, Y., Du, H., and Gong, Q. (2017). LncRNA expression in the spinal cord modulated by minocycline in a mouse model of spared nerve injury. J. Pain Res. 10: 2503–2514, https://doi.org/10.2147/JPR.S147055.Search in Google Scholar PubMed PubMed Central

Liu, Z., Song, Z., Guo, S., He, J., Wang, S., Zhu, J., Yang, H., and Liu, J. (2019). CXCL12/CXCR4 signaling contributes to neuropathic pain via central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neurosci. Ther. 25: 922–936, https://doi.org/10.1111/cns.13128.Search in Google Scholar PubMed PubMed Central

Lu, K., Zhao, L., Zhang, Y., Yang, F., Zhang, H., Wang, J., Li, B., Ji, G., Yu, J., and Ma, H. (2022). Bupivacaine reduces GlyT1 expression by potentiating the p-AMPKα/BDNF signalling pathway in spinal astrocytes of rats. Sci. Rep. 12: 1378, https://doi.org/10.1038/s41598-022-05478-3.Search in Google Scholar PubMed PubMed Central

Luo, X., Tai, W.L., Sun, L., Pan, Z., Xia, Z., Chung, S.K., and Cheung, C.W. (2016). Crosstalk between astrocytic CXCL12 and microglial CXCR4 contributes to the development of neuropathic pain. Mol. Pain 12: 174480691663638, https://doi.org/10.1177/1744806916636385.Search in Google Scholar PubMed PubMed Central

Ma, Y., Liu, Y., and Jiang, Z. (2020). CircRNAs: a new perspective of biomarkers in the nervous system. Biomed. Pharmacother. 128: 110251, https://doi.org/10.1016/j.biopha.2020.110251.Search in Google Scholar PubMed

Masri, R. and Keller, A. (2012). Chronic pain following spinal cord injury. In: Jandial, R. and Chen, M.Y. (Eds.), Regenerative biology of the spine and spinal cord. Springer New York, New York, NY, pp. 74–88.10.1007/978-1-4614-4090-1_5Search in Google Scholar PubMed PubMed Central

Mauceri, D. (2022). Role of epigenetic mechanisms in chronic pain. Cells 11: 2613, https://doi.org/10.3390/cells11162613.Search in Google Scholar PubMed PubMed Central

Mazaleuskaya, L.L., Muzykantov, V.R., and FitzGerald, G.A. (2021). Nanotherapeutic-directed approaches to analgesia. Trends Pharmacol. Sci. 42: 527–550, https://doi.org/10.1016/j.tips.2021.03.007.Search in Google Scholar PubMed PubMed Central

Mazzone, G.L. and Nistri, A. (2019). Modulation of extrasynaptic GABAergic receptor activity influences glutamate release and neuronal survival following excitotoxic damage to mouse spinal cord neurons. Neurochem. Int. 128: 175–185, https://doi.org/10.1016/j.neuint.2019.04.018.Search in Google Scholar PubMed

Mazzone, G.L., Mohammadshirazi, A., Aquino, J.B., Nistri, A., and Taccola, G. (2021). GABAergic mechanisms can redress the tilted balance between excitation and inhibition in damaged spinal networks. Mol. Neurobiol. 58: 3769–3786, https://doi.org/10.1007/s12035-021-02370-5.Search in Google Scholar PubMed PubMed Central

Miguel, C.A., Noya-Riobó, M.V., Mazzone, G.L., Villar, M.J., and Coronel, M.F. (2021). Antioxidant, anti-inflammatory and neuroprotective actions of resveratrol after experimental nervous system insults. Special focus on the molecular mechanisms involved. Neurochem. Int. 150: 105188, https://doi.org/10.1016/j.neuint.2021.105188.Search in Google Scholar PubMed

Miyakoshi, N., Suda, K., Kudo, D., Sakai, H., Nakagawa, Y., Mikami, Y., Suzuki, S., Tokioka, T., Tokuhiro, A., Takei, H., et al.. (2021). A nationwide survey on the incidence and characteristics of traumatic spinal cord injury in Japan in 2018. Spinal Cord 59: 626–634, https://doi.org/10.1038/s41393-020-00533-0.Search in Google Scholar PubMed

Morita, K., Motoyama, N., Kitayama, T., Morioka, N., Kifune, K., and Dohi, T. (2008). Spinal antiallodynia action of Glycine transporter inhibitors in neuropathic pain models in mice. J. Pharmacol. Exp. Therapeut. 326: 633–645, https://doi.org/10.1124/jpet.108.136267.Search in Google Scholar PubMed

Nakagawa, T. and Kaneko, S. (2010). Spinal astrocytes as therapeutic targets for pathological pain. J. Pharmacol. Sci. 114: 347–353, https://doi.org/10.1254/jphs.10R04CP.Search in Google Scholar

Napier, I.A., Mohammadi, S.A., and Christie, M.J. (2012). Glutamate transporter dysfunction associated with nerve injury-induced pain in mice. J. Neurophysiol. 107: 649–657, https://doi.org/10.1152/jn.00763.2011.Search in Google Scholar PubMed

Nikolenko, V.N., Shelomentseva, E.M., Tsvetkova, M.M., Abdeeva, E.I., Giller, D.B., Babayeva, J.V., Achkasov, E.E., Gavryushova, L.V., and Sinelnikov, M.Y. (2022). Nociceptors: their role in body’s defenses, tissue specific variations and anatomical update. J. Pain Res. 15: 867–877, https://doi.org/10.2147/JPR.S348324.Search in Google Scholar PubMed PubMed Central

Ohnishi, T., Iwasaki, N., and Sudo, H. (2022). Causes of and molecular targets for the treatment of intervertebral disc degeneration: a review. Cells 11: 394, https://doi.org/10.3390/cells11030394.Search in Google Scholar PubMed PubMed Central

Ozata, D.M., Gainetdinov, I., Zoch, A., O’Carroll, D., and Zamore, P.D. (2019). PIWI-interacting RNAs: small RNAs with big functions. Nat. Rev. Genet. 20: 89–108, https://doi.org/10.1038/s41576-018-0073-3.Search in Google Scholar PubMed

Palazzo, E., Marabese, I., Luongo, L., Guida, F., de Novellis, V., and Maione, S. (2017). Nociception modulation by supraspinal group III metabotropic glutamate receptors. J. Neurochem. 141: 507–519, https://doi.org/10.1111/jnc.13725.Search in Google Scholar PubMed

Pan, B., Zhou, H.-X., Liu, Y., Yan, J.-Y., Wang, Y., Yao, X., Deng, Y.-Q., Chen, S.-Y., Lu, L., Wei, Z.-J., et al.. (2017). Time-dependent differential expression of long non-coding RNAs following peripheral nerve injury. Int. J. Mol. Med. 39: 1381–1392, https://doi.org/10.3892/ijmm.2017.2963.Search in Google Scholar PubMed PubMed Central

Panda, A.C. (2018). Circular RNAs act as miRNA sponges. In: Xiao, J. (Ed.), Circular RNAs. Springer Singapore, Singapore, pp. 67–79.10.1007/978-981-13-1426-1_6Search in Google Scholar PubMed

Park, J. and Luo, Z.D. (2010). Calcium channel functions in pain processing. Channels 4: 510–517, https://doi.org/10.4161/chan.4.6.12869.Search in Google Scholar PubMed PubMed Central

Park, E., Velumian, A.A., and Fehlings, M.G. (2004). The role of excitotoxicity in secondary mechanisms of spinal cord injury: a review with an emphasis on the implications for white matter degeneration. J. Neurotrauma 21: 754–774, https://doi.org/10.1089/0897715041269641.Search in Google Scholar PubMed

Pavlov, V.A. and Tracey, K.J. (2005). The cholinergic anti-inflammatory pathway. Brain Behav. Immun. 19: 493–499, https://doi.org/10.1016/j.bbi.2005.03.015.Search in Google Scholar PubMed

Peirs, C., Williams, S.-P.G., Zhao, X., Walsh, C.E., Gedeon, J.Y., Cagle, N.E., Goldring, A.C., Hioki, H., Liu, Z., Marell, P.S., et al.. (2015). Dorsal horn circuits for persistent mechanical pain. Neuron 87: 797–812, https://doi.org/10.1016/j.neuron.2015.07.029.Search in Google Scholar PubMed PubMed Central

Piwecka, M., Glažar, P., Hernandez-Miranda, L.R., Memczak, S., Wolf, S.A., Rybak-Wolf, A., Filipchyk, A., Klironomos, F., Cerda Jara, C.A., Fenske, P., et al.. (2017). Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 357: eaam8526, https://doi.org/10.1126/science.aam8526.Search in Google Scholar PubMed

Prescott, S.A., Ma, Q., and De Koninck, Y. (2014). Normal and abnormal coding of somatosensory stimuli causing pain. Nat. Neurosci. 17: 183–191, https://doi.org/10.1038/nn.3629.Search in Google Scholar PubMed PubMed Central

Qu, X.-X., Cai, J., Li, M.-J., Chi, Y.-N., Liao, F.-F., Liu, F.-Y., Wan, Y., Han, J.-S., and Xing, G.-G. (2009). Role of the spinal cord NR2B-containing NMDA receptors in the development of neuropathic pain. Exp. Neurol. 215: 298–307, https://doi.org/10.1016/j.expneurol.2008.10.018.Search in Google Scholar PubMed

Quemener, A.M., Centomo, M.L., Sax, S.L., and Panella, R. (2022). Small drugs, huge impact: the extraordinary impact of antisense oligonucleotides in research and drug development. Molecules 27: 536, https://doi.org/10.3390/molecules27020536.Search in Google Scholar PubMed PubMed Central

Qureshi, I.A. and Mehler, M.F. (2013). Long non-coding RNAs: novel targets for nervous system disease diagnosis and therapy. Neurotherapeutics 10: 632–646, https://doi.org/10.1007/s13311-013-0199-0.Search in Google Scholar PubMed PubMed Central

Ray, P., Torck, A., Quigley, L., Wangzhou, A., Neiman, M., Rao, C., Lam, T., Kim, J.-Y., Kim, T.H., Zhang, M.Q., et al.. (2018). Comparative transcriptome profiling of the human and mouse dorsal root ganglia: an RNA-seq-based resource for pain and sensory neuroscience research. Pain 159: 1325–1345, https://doi.org/10.1097/j.pain.0000000000001217.Search in Google Scholar PubMed PubMed Central

Roganović, J. and Petrović, N. (2022). Clinical perspectives of non-coding RNA in oral inflammatory diseases and neuropathic pain: a narrative review. Int. J. Mol. Sci. 23: 8278, https://doi.org/10.3390/ijms23158278.Search in Google Scholar PubMed PubMed Central

Rybak-Wolf, A., Stottmeister, C., Glažar, P., Jens, M., Pino, N., Giusti, S., Hanan, M., Behm, M., Bartok, O., Ashwal-Fluss, R., et al.. (2015). Circular RNAs in the Mammalian brain are highly abundant, conserved, and dynamically expressed. Mol. Cell 58: 870–885, https://doi.org/10.1016/j.molcel.2015.03.027.Search in Google Scholar PubMed

Salami, R., Salami, M., Mafi, A., Vakili, O., and Asemi, Z. (2022). Circular RNAs and glioblastoma multiforme: focus on molecular mechanisms. Cell Commun. Signal. 20: 13, https://doi.org/10.1186/s12964-021-00809-9.Search in Google Scholar PubMed PubMed Central

Sámano, C., Mladinic, M., and Mazzone, G.L. (2021). Circular RNAs: the novel actors in pathophysiology of spinal cord injury. Front. Integr. Neurosci. 15: 758340, https://doi.org/10.3389/fnint.2021.758340.Search in Google Scholar PubMed PubMed Central

Santa-Cecília, F.V., Ferreira, D.W., Guimaraes, R.M., Cecilio, N.T., Fonseca, M.M., Lopes, A.H., Davoli-Ferreira, M., Kusuda, R., Souza, G.R., Nachbur, U., et al.. (2019). The NOD2 signaling in peripheral macrophages contributes to neuropathic pain development. Pain 160: 102–116, https://doi.org/10.1097/j.pain.0000000000001383.Search in Google Scholar PubMed

Shen, W., Hu, X.-M., Liu, Y.-N., Han, Y., Chen, L.-P., Wang, C.-C., and Song, C. (2014). CXCL12 in astrocytes contributes to bone cancer pain through CXCR4-mediated neuronal sensitization and glial activation in rat spinal cord. J. Neuroinflammation 11: 75, https://doi.org/10.1186/1742-2094-11-75.Search in Google Scholar PubMed PubMed Central

Shin, D., Lee, W., Lee, J.H., and Bang, D. (2019). Multiplexed single-cell RNA-seq via transient barcoding for simultaneous expression profiling of various drug perturbations. Sci. Adv. 5: eaav2249, https://doi.org/10.1126/sciadv.aav2249.Search in Google Scholar PubMed PubMed Central

Song, G., Yang, Z., Guo, J., Zheng, Y., Su, X., and Wang, X. (2020). Interactions among lncRNAs/circRNAs, miRNAs, and mRNAs in neuropathic pain. Neurother. J. Am. Soc. Exp. Neurother. 17: 917–931, https://doi.org/10.1007/s13311-020-00881-y.Search in Google Scholar PubMed PubMed Central

St. Laurent, G., Wahlestedt, C., and Kapranov, P. (2015). The Landscape of long noncoding RNA classification. Trends Genet. 31: 239–251, https://doi.org/10.1016/j.tig.2015.03.007.Search in Google Scholar PubMed PubMed Central

Su, J.-F., Concilla, A., Zhang, D., Zhao, F., Shen, F.-F., Zhang, H., and Zhou, F.-Y. (2020). PIWI-interacting RNAs: mitochondria-based biogenesis and functions in cancer. Genes Dis. 8: 603–622, https://doi.org/10.1016/j.gendis.2020.09.006.Search in Google Scholar PubMed PubMed Central

Tang, S., Zhou, J., Jing, H., Liao, M., Lin, S., Huang, Z., Huang, T., Zhong, J., and Wang, H. (2019). Functional roles of lncRNAs and its potential mechanisms in neuropathic pain. Clin. Epigenet. 11: 78, https://doi.org/10.1186/s13148-019-0671-8.Search in Google Scholar PubMed PubMed Central

Tao, Y.-X., Gu, J., and Stephens, R.L. (2005). Role of spinal cord glutamate transporter during normal sensory transmission and pathological pain states. Mol. Pain 1: 30, https://doi.org/10.1186/1744-8069-1-30.Search in Google Scholar PubMed PubMed Central

Thomas, A.X., Riviello, J.J., Davila-Williams, D., Thomas, S.P., Erklauer, J.C., Bauer, D.F., and Cokley, J.A. (2022). Pharmacologic and acute management of spinal cord injury in adults and children. Curr. Treat. Options Neurol. 24: 285–304, https://doi.org/10.1007/s11940-022-00720-9.Search in Google Scholar PubMed PubMed Central

Tian, M.-M., Li, Y.-X., Liu, S., Zhu, C.-H., Lan, X.-B., Du, J., Ma, L., Yang, J.-M., Zheng, P., Yu, J.-Q., et al.. (2021). Glycosides for peripheral neuropathic pain: a potential medicinal components. Molecules 27: 255, https://doi.org/10.3390/molecules27010255.Search in Google Scholar PubMed PubMed Central

Tong, D., Zhao, Y., Tang, Y., Ma, J., Wang, Z., and Li, C. (2021). Circ-Usp10 promotes microglial activation and induces neuronal death by targeting miRNA-152-5p/CD84. Bioengineered 12: 10812–10822, https://doi.org/10.1080/21655979.2021.2004362.Search in Google Scholar PubMed PubMed Central

Tsuda, M. (2016). Microglia in the spinal cord and neuropathic pain. J. Diabetes Invest. 7: 17–26, https://doi.org/10.1111/jdi.12379.Search in Google Scholar PubMed PubMed Central

von Hehn, C.A., Baron, R., and Woolf, C.J. (2012). Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 73: 638–652, https://doi.org/10.1016/j.neuron.2012.02.008.Search in Google Scholar PubMed PubMed Central

Wang, K., Wang, T., Gao, X.-Q., Chen, X.-Z., Wang, F., and Zhou, L.-Y. (2021a). Emerging functions of PIWI-interacting RNAs in diseases. J. Cell Mol. Med. 25: 4893–4901, https://doi.org/10.1111/jcmm.16466.Search in Google Scholar PubMed PubMed Central

Wang, L., Luo, T., Bao, Z., Li, Y., and Bu, W. (2018). Intrathecal circHIPK3 shRNA alleviates neuropathic pain in diabetic rats. Biochem. Biophys. Res. Commun. 505: 644–650, https://doi.org/10.1016/j.bbrc.2018.09.158.Search in Google Scholar PubMed

Wang, N., Zhang, Y.-H., Wang, J.-Y., and Luo, F. (2021b). Current understanding of the involvement of the insular cortex in neuropathic pain: a narrative review. Int. J. Mol. Sci. 22: 2648, https://doi.org/10.3390/ijms22052648.Search in Google Scholar PubMed PubMed Central

Wang, Y.-Z., Wu, C.-C., and Wang, X.-Q. (2021c). Bibliometric study of pain after spinal cord injury. Neural Plast. 2021: 1–15, https://doi.org/10.1155/2021/6634644.Search in Google Scholar PubMed PubMed Central

Wei, M., Li, L., Zhang, Y., Zhang, M., and Su, Z. (2020). Downregulated circular RNA zRANB1 mediates Wnt5a/β-catenin signaling to promote neuropathic pain via miR-24-3p/LPAR3 axis in CCI rat models. Gene 761: 145038, https://doi.org/10.1016/j.gene.2020.145038.Search in Google Scholar PubMed

Widerström-Noga, E. (2017). Neuropathic pain and spinal cord injury: phenotypes and pharmacological management. Drugs 77: 967–984, https://doi.org/10.1007/s40265-017-0747-8.Search in Google Scholar PubMed

Wiffen, P.J., Derry, S., Bell, R.F., Rice, A.S., Tölle, T.R., Phillips, T., and Moore, R.A. (2017). Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2020: CD007938, https://doi.org/10.1002/14651858.CD007938.pub4.Search in Google Scholar PubMed PubMed Central

Willemen, H.L.D.M., Eijkelkamp, N., Wang, H., Dantzer, R., Dorn, G.W., Kelley, K.W., Heijnen, C.J., and Kavelaars, A. (2010). Microglial/macrophage GRK2 determines duration of peripheral IL-1β-induced hyperalgesia: contribution of spinal cord CX3CR1, p38 and IL-1 signaling. Pain 150: 550–560, https://doi.org/10.1016/j.pain.2010.06.015.Search in Google Scholar PubMed PubMed Central

Winkle, M., El-Daly, S.M., Fabbri, M., and Calin, G.A. (2021). Noncoding RNA therapeutics—challenges and potential solutions. Nat. Rev. Drug Discov. 20: 629–651, https://doi.org/10.1038/s41573-021-00219-z.Search in Google Scholar PubMed PubMed Central

Witiw, C.D. and Fehlings, M.G. (2015). Acute spinal cord injury. J. Spinal Disord. Tech. 28: 202–210, https://doi.org/10.1097/BSD.0000000000000287.Search in Google Scholar PubMed

Wu, P.-C. and Kao, L.-S. (2016). Calcium regulation in mouse mesencephalic neurons—differential roles of Na+/Ca2+ exchanger, mitochondria and endoplasmic reticulum. Cell Calcium 59: 299–311, https://doi.org/10.1016/j.ceca.2016.03.008.Search in Google Scholar PubMed

Wu, Q., Yue, J., Lin, L., Yu, X., Zhou, Y., Ying, X., Chen, X., Tu, W., Lou, X., Yang, G., et al.. (2021). Electroacupuncture may alleviate neuropathic pain via suppressing P2X7R expression. Mol. Pain 17: 174480692199765, https://doi.org/10.1177/1744806921997654.Search in Google Scholar PubMed PubMed Central

Wu, W., Ji, X., and Zhao, Y. (2019). Emerging roles of long non-coding RNAs in chronic neuropathic pain. Front. Neurosci. 13: 1097, https://doi.org/10.3389/fnins.2019.01097.Search in Google Scholar PubMed PubMed Central

Xia, L., Ke, C., and Lu, J. (2018). NEAT1 contributes to neuropathic pain development through targeting miR-381/HMGB1 axis in CCI rat models. J. Cell. Physiol. 233: 7103–7111, https://doi.org/10.1002/jcp.26526.Search in Google Scholar PubMed

Xiang, Q., Kang, L., Wang, J., Liao, Z., Song, Y., Zhao, K., Wang, K., Yang, C., and Zhang, Y. (2020). CircRNA-CIDN mitigated compression loading-induced damage in human nucleus pulposus cells via miR-34a-5p/SIRT1 axis. EBioMedicine 53: 102679, https://doi.org/10.1016/j.ebiom.2020.102679.Search in Google Scholar PubMed PubMed Central

Xin, Y., Song, X., and Ge, Q. (2021). Circular RNA SMEK1 promotes neuropathic pain in rats through targeting microRNA-216a-5p to mediate Thioredoxin Interacting Protein (TXNIP) expression. Bioengineered 12: 5540–5551, https://doi.org/10.1080/21655979.2021.1965811.Search in Google Scholar PubMed PubMed Central

Xu, D., Ma, X., Sun, C., Han, J., Zhou, C., Chan, M.T.V., and Wu, W.K.K. (2021a). Emerging roles of circular RNAs in neuropathic pain. Cell Prolif 54: e13139, https://doi.org/10.1111/cpr.13139.Search in Google Scholar PubMed PubMed Central

Xu, S., Dong, H., Zhao, Y., and Feng, W. (2021b). Differential expression of long non-coding RNAs and their role in rodent neuropathic pain models. J. Pain Res. 14: 3935–3950, https://doi.org/10.2147/JPR.S344339.Search in Google Scholar PubMed PubMed Central

Yu, X., Liu, H., Hamel, K.A., Morvan, M.G., Yu, S., Leff, J., Guan, Z., Braz, J.M., and Basbaum, A.I. (2020). Dorsal root ganglion macrophages contribute to both the initiation and persistence of neuropathic pain. Nat. Commun. 11: 264, https://doi.org/10.1038/s41467-019-13839-2.Search in Google Scholar PubMed PubMed Central

Zhang, S.-B., Lin, S.-Y., Liu, M., Liu, C.-C., Ding, H.-H., Sun, Y., Ma, C., Guo, R.-X., Lv, Y.-Y., Wu, S.-L., et al.. (2019). CircAnks1a in the spinal cord regulates hypersensitivity in a rodent model of neuropathic pain. Nat. Commun. 10: 4119, https://doi.org/10.1038/s41467-019-12049-0.Search in Google Scholar PubMed PubMed Central

Zhang, Y., Gao, T., Li, X., Wen, C.-C., Yan, X.-T., Peng, C., and Xiao, Y. (2021). Circ_0005075 targeting miR-151a-3p promotes neuropathic pain in CCI rats via inducing NOTCH2 expression. Gene 767: 145079, https://doi.org/10.1016/j.gene.2020.145079.Search in Google Scholar PubMed

Zhang, Z., Yang, T., and Xiao, J. (2018). Circular RNAs: promising biomarkers for human diseases. EBioMedicine 34: 267–274, https://doi.org/10.1016/j.ebiom.2018.07.036.Search in Google Scholar PubMed PubMed Central

Zheng, Y.-L., Guo, J.-B., Song, G., Yang, Z., Su, X., Chen, P.-J., and Wang, X.-Q. (2022). The role of circular RNAs in neuropathic pain. Neurosci. Biobehav. Rev. 132: 968–975, https://doi.org/10.1016/j.neubiorev.2021.10.048.Search in Google Scholar PubMed

Zhou, J., Xiong, Q., Chen, H., Yang, C., and Fan, Y. (2017). Identification of the spinal expression profile of non-coding RNAs involved in neuropathic pain following spared nerve injury by sequence analysis. Front. Mol. Neurosci. 10: 91, https://doi.org/10.3389/fnmol.2017.00091.Search in Google Scholar PubMed PubMed Central

Zhou, Z., Niu, Y., Huang, G., Lu, J., Chen, A., and Zhu, L. (2018). Silencing of circRNA.2837 plays a protective role in sciatic nerve injury by sponging the miR-34 family via regulating neuronal autophagy. Mol. Ther. Nucleic Acids 12: 718–729, https://doi.org/10.1016/j.omtn.2018.07.011.Search in Google Scholar PubMed PubMed Central

Zhuang, Z.-Y., Gerner, P., Woolf, C.J., and Ji, R.-R. (2005). ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 114: 149–159, https://doi.org/10.1016/j.pain.2004.12.022.Search in Google Scholar PubMed

Zhuo, M. (2007). Neuronal mechanism for neuropathic pain. Mol. Pain 3: 14, https://doi.org/10.1186/1744-8069-3-14.Search in Google Scholar PubMed PubMed Central

Zhuo, M., Wu, G., and Wu, L.-J. (2011). Neuronal and microglial mechanisms of neuropathic pain. Mol. Brain 4: 31, https://doi.org/10.1186/1756-6606-4-31.Search in Google Scholar PubMed PubMed Central

Zorzopulos, J., Opal, S.M., Hernando-Insúa, A., Rodriguez, J.M., Elías, F., Fló, J., López, R.A., Chasseing, N.A., Lux-Lantos, V.A., Coronel, M.F., et al.. (2017). Immunomodulatory oligonucleotide IMT504: effects on mesenchymal stem cells as a first-in-class immunoprotective/immunoregenerative therapy. World J. Stem Cell. 9: 45, https://doi.org/10.4252/wjsc.v9.i3.45.Search in Google Scholar PubMed PubMed Central

Received: 2022-07-22
Accepted: 2022-10-19
Published Online: 2022-11-10
Published in Print: 2023-08-28

© 2022 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 18.5.2024 from https://www.degruyter.com/document/doi/10.1515/revneuro-2022-0089/html
Scroll to top button