Abstract
Neuropathic pain is a widespread clinical issue caused by somatosensory nervous system damage, affecting numerous individuals. It poses considerable economic and public health challenges, and managing it can be challenging due to unclear underlying mechanisms. Nevertheless, emerging evidence suggests that neurogenic inflammation and neuroinflammation play a role in developing pain patterns. Emerging evidence suggests that neurogenic inflammation and neuroinflammation play significant roles in developing neuropathic pain within the nervous system. Increased/decreased miRNA expression patterns could affect the progression of neuropathic and inflammatory pain by controlling nerve regeneration, neuroinflammation, and the expression of abnormal ion channels. However, our limited knowledge of miRNA targets hinders a complete grasp of miRNA’s functions. Meanwhile, exploring exosomal miRNA, a recently uncovered role, has significantly advanced our comprehension of neuropathic pain’s pathophysiology in recent times. In this review, we present a comprehensive overview of the latest miRNA studies and explore the possible ways miRNAs might play a role in the development of neuropathic pain.
Keywords: Micro RNA, neuropathic, pain
Introduction
Neuropathic pain, a condition arising from somatosensory nervous system damage, is a prevalent global clinical issue impacting numerous individuals worldwide . 1 It poses substantial public health and economic challenges and proves challenging to manage due to unclear underlying mechanisms. However, emerging evidence points to the involvement of neuroinflammation and neurogenic inflammation in the development of pain. 2 Furthermore, a growing body of evidence indicates that the stimulation of neuroinflammation and neurogenic inflammation, in the peripheral and central nervous systems, plays a critical role in the prolonged existence of pain.3,4 Previous studies have investigated the possibility that chronic neuropathic pain could stem from an inflammatory response within the nervous system triggered by nerve damage. 5 Recent research findings suggest that microRNAs (miRNAs) exhibit important anti-inflammatory effects, influencing pathogenesis in various contexts, including tumors, inflammation, cardiovascular and respiratory diseases.6–9 However, the exact pathogenesis of neuropathic pain remains unclear. Newer investigations employing microarray technologies have uncovered the significant involvement of miRNAs in the emergence and advancement of neuropathic pain. Their regulatory impact extends to various aspects, including neuronal plasticity, excitability, ion channels, synaptic plasticity, and neuroinflammation. The gathered evidence indicates that changes in miRNA expression patterns may be critical players in the development of neuropathic pain. Nevertheless, the lack of understanding regarding miRNA targets hinders a comprehensive grasp of miRNAs’ biological functions in this context. This review aims to shed light on miRNA expression profiling studies related to neuropathic pain and explores specific miRNAs' molecular mechanisms that significantly contribute to neuropathic pain pathogenesis. Furthermore, we discuss the potential prognostic, diagnostic, and therapeutic implications of these miRNAs in the clinical management of neuropathic and neurogenic pain.
The roles of miRNAs
MicroRNAs (miRNAs) are prevalent in mammalian cells and are crucial in regulating approximately 30% of human genes. In the human genome, there are over 1800 miRNAs that target around 60% of human mRNAs. 10 Mutations in miRNA and related pathway genes have been linked to various human disorders, including neuropathic pain. MicroRNAs are abundantly distributed in both the central and peripheral nervous systems, which contain components related to pain processing. Although miRNAs do not directly engage in synthesizing peptides, they belong to the group of noncoding RNAs that hold substantial sway over miRNA stability and the translation of proteins. 11 miRNAs exert their influence on gene expression after transcription by binding to the 3′UTR of target mRNAs, resulting in a notable decrease (84%) in the production of proteins. 12 It is noteworthy that a single miRNA can target multiple mRNA transcripts, and multiple miRNAs can act on a single mRNA molecule simultaneously.12,13 There is a widely accepted consensus regarding the critical roles miRNAs play in regulating various essential biological processes, such as early development, cell proliferation, apoptosis, lipid metabolism, cell differentiation, and disease progression. 14 Research conducted in experimental models has indicated that changes in miRNA expression patterns could impact the development of a wide range of conditions, such as cardiovascular diseases, cerebrovascular diseases, and neurodegenerative disorders.15,16
Regulatory mechanisms of miRNAs in neuropathic pain
Several research investigations showed notable irregularities in miRNA expression in animals after experiencing peripheral nerve injury. Some of the miRNAs that have been discovered are linked to neuroinflammation, abnormal ion channel expression, and nerve regeneration, suggesting that altered miRNA expression could play a role in the development of neuropathic pain and might have the potential as useful diagnostic markers, enhancing the categorization of neuropathic pain. Nevertheless, the lack of information regarding miRNA targets poses a challenge to fully understanding the functions of miRNAs. In this section, we present an extensive summary of the existing understanding of miRNAs and investigate how they may play a role in neuropathic pain (Table 1).
Table 1.
The role of Micro RNAs in neuropathic pain.
| Mechanism | Micro RNA | Target | Effect | Reference |
| Neuropathic pain | miR-128-3p ↑ | ZEB1 ↓ | Eased the development of neuropathic pain. | 17 |
| miR-23a ↑ | CXCR4/TXNIP/NLRP3 ↓ | Suppressed the formation of neuropathic pain. | 18 | |
| miR-221 ↓ | SOCS1 ↑ | Mitigated neuropathic pain and neuroinflammation. | 19 | |
| miR-22 ↑ | Mtf1 ↑ | Facilitated the growth and sustenance of pain associated with inflammation. | 20 | |
| miR-381 ↑ | HMGB1 and CXCR4 ↓ | Hindered the progression of neuropathic pain. | 18 | |
| miR-216a-5p ↑ | KDM3A ↓ | Relieved rats of neuropathic pain. | 21 | |
| miR-150 ↑ | ZEB1 ↓ | Suppressed neuropathic pain in vivo. | 22,23 | |
| miR-155 ↓ | SOCS1 ↑ | Reduced the intensity of neuropathic pain. | 24 | |
| miR-144 ↑ | RASA1 ↓ | Aided in preventing the progression of neuropathic pain. | 17 | |
| miR-362-3p ↑ | BAMBI ↓ | The progression of neuropathic pain was repressed. | 25 | |
| miR-221 ↓ | SOCS3 ↑ | Diminished discomfort and decreased inflammatory marker expression. | 26 | |
| miR-93 ↑ | STAT3 ↓ | Suppressed the formation of neuropathic pain in rats with chronic constriction injury. | 27 | |
| miR-155 ↓ | TRPA1 ↓ | Dampened the neuropathic pain triggered by OXL. | 28 | |
| miR-183 ↑ | TGF-α/CCL2/CCR2 ↓ | The pain associated with osteoarthritis was improved. | 29 | |
| miR-140 ↑ | S1PR1 ↓ | Inhibited neuropathic pain induced by chronic constriction injury. | 21 | |
| miR-28-5p ↑ | ZEB1 ↓ | Diminished sensations of neuropathic pain. | 30 | |
| miR-429 ↑ | ZEB1 ↓ | Diminished the progression of neuropathic pain in vivo. | 22,23 | |
| miR-136 ↑ | ZEB1 ↓ | Prevented the advancement of neuropathic pain. | 31 | |
| Infiltration of immune cells | miR-214-3p ↑ | CSF1 ↓ | Diminished the neuroinflammatory response and pain-related behavior. | 32 |
| miR-146a-5p ↑ | IRAK1/TRAF6 ↓ | Prevented the formation of neuropathic pain caused by chronic constriction injury. | 33 | |
| miR-590-3p ↑ | RAP1A ↓ | A decrease in T cell infiltration resulted in the obstruction of diabetic peripheral neuropathy pain (DPNP). | 34 | |
| miR-135s ↑ | KLF4 ↓ | Promoted the regrowth of retinal ganglion cell axons following optic nerve damage in mature mice. | 35 | |
| miR-155 ↓ | SPRR1A ↑ | Improved the viability of neurons and facilitated the extension of axons. | 36 | |
| miR-192-5p ↑ | XIAP ↑ | Reduced the programmed cell death of nerve cells and supported the recovery and regrowth of peripheral nerves following injury. | 37 | |
| miR-125b ↑ | JAK/STAT ↓ | Facilitated the restoration and growth process after damage to the spinal cord. | 38 | |
| miR-210 ↑ | EFNA3 ↓ | Stimulated the regrowth of sensory axons while impeding programmed cell death. | 39 | |
| miR-138 ↓ | SIRT1 ↑ | Facilitated the regrowth of axons in mammals. | 40 | |
| miR-21 ↑ | EGFR ↑ | Improved the regrowth of axons following optic nerve crush. | 41 | |
| miR-21 ↑ | PTEN ↓ | Stimulated neurite growth. | 42 | |
| miR-21 ↑ | TGFβI/TIMP3/EPHA4 ↓ | Facilitated the multiplication of schwann cells and the regrowth of axons. | 43 | |
| miR let-7 ↑ | Ntn1 ↓ | Diminished the extension of axons. | 44 | |
| miR-9 ↑ | Dcc ↓ | Diminished the extension of axons. | 44 | |
| miR-199a-3p ↑ | mTOR ↓ | Weakened neurite growth. | 42 | |
| miR-26a ↑ | GSK3β/Smad1 ↓ | Aided the regrowth of axons in vivo. | 45 | |
| miR-9 ↑ | FoxP1 ↓ | Inhibited axon regeneration in vitro and in vivo. | 46 | |
| miR-455-5p ↓ | GSK3β/Tau ↑ | Facilitated the expansion and renewal of axons. | 47 | |
| miR-19a ↑ | PTEN ↑ | Encouraged the regrowth of axons following optic nerve injury in mature mice. | 48 | |
| Voltage-gated potassium channels | miR-137 ↓ | Kv1.2 ↑ | Relieved sensitivity to touch and extreme sensitivity to heat. | 49 |
| miR-183-5P ↑ | TREK-1 ↓ | Effectively improved the condition of neuropathic pain. | 50 | |
| miR-17-92 ↓ | Multiple voltage-gated potassium channels ↑ | Reduced the hypersensitivity to touch caused by nerve damage. | 51 | |
| Voltage-gated calcium channels | miR-103 ↑ | Cav1.2-LTC ↓ | Effectively alleviate discomfort. | 52 |
| Voltage-gated sodium channel | miR-219 ↑ | CaMKIIγ ↓ | Blocked and reverted neuropathic pain and increased sensitivity of spinal neurons triggered by complete Freund’s adjuvant. | 53 |
| miR-183 ↑ | α2δ-1 and α2δ-2 | Halted the increase in inherent mechanical sensitivity in pain receptors. | 54 | |
| miR-32-5p ↑ | Cav3.2 ↓ | Reversed mechanical allodynia. | 55 | |
| miR-124a ↑ | MeCP2 ↓ | Reduced the perception of pain caused by inflammation. | 56 | |
| miR-30b ↑ | Nav1.7 ↓ | Alleviated neuropathic pain. | 57 | |
| miR-7a ↑ | β2 subunit ↓ | Inhibited the sensation of neuropathic pain. | 51 | |
| miR-30b ↑ | Nav1.3 ↓ | Inhibited the sensation of neuropathic pain. | 58 | |
| miR-183 ↑ | Nav1.3/Nav1.7/Nav1.8 ↓ | Blocked the presence of pain-associated elements and improved the pain caused by osteoarthritis. | 29 | |
| miR-384-5p ↑ | SCN3A ↓ | Markedly reduced heightened sensitivity to touch and extreme sensitivity to heat in rats with chronic constriction injury. | 59 | |
| miR-182 ↑ | Nav1.7 ↓ | Relieved neuropathic pain caused by spared nerve injury. | 60 | |
| miR-96 ↑ | Nav1.3 ↓ | Eased the discomfort of neuropathic pain. | 61 | |
| miR-30b-5p ↑ | Nav1.6 ↓ | Attenuated pain. | 62 |
PTEN: phosphatase and tensin homolog; DPNP: diabetic peripheral neuropathic pain; KDM3A: lysine-specific demethylase 3A; CSF1: Colony-stimulating factor 1; TGFβI: transforming growth factor-beta-induced protein; TRAF6: Tumor necrosis factor receptor-associated factor 6; TIMP3: tissue inhibitor of metalloproteinases 3; HMGB1: high mobility group box 1; FoxP1: forkhead box protein P1; CXCR4: Chemokine CXC receptor 4; GSK3β: glycogen synthase kinase 3β; TGF-α: Tumor necrosis factor α; KLF4: kruppel-like factor 4; CCL2: CC chemokine ligand 2; IRAK: IL-1R-associated kinase; CCR2: CC chemokine receptor type-2; MeCP2: Methyl-CpG binding protein 2; ZEB1: Zinc finger E-box-binding homeobox 1; TXNIP: Thioredoxin interacting protein; NLRP3: NOD-like receptor protein 3; Dcc: deleted in colorectal cancer; S1PR1: Sphingosine-1-phosphate receptor 1; Ntn1: netrin-1; RASA1: RAS P21 protein activator 1; RAP1A: Ras-related protein 1A; STAT: signal transducer and activator of transcription; Mtf1: Mitochondrial Transcription Factor 1; BAMBI: bone morphogenetic protein and activin membrane-bound inhibitor; OXL: Oxaliplatin; XIAP: X-linked inhibitor of apoptosis protein; EFNA3: ephrin-A3; SPRR1A: small proline-rich repeat protein 1A; EPHA4: erythropoietin-producing human hepatocellular receptor A4; SOCS: Suppressor of cytokine signaling; mTOR: mechanistic target of rapamycin; TRPA1: transient receptor potential cation channel subfamily A member 1; SIRT1: Sirtuin 1; Nav: voltage-gated sodium channels; JAK: Janus kinase gene; Kv: voltage-gated potassium channels; TREK-1: TWIK-related K channel 1; EGFR: epidermal growth factor receptor; Cav1.2-LTC: Cav1.2-comprising L-type calcium channel; CaMKII: calmodulin (CaM)-activated kinase II.
Regulation of neuroinflammation in neuropathic pain enhancement
MiRNAs have shown their involvement in nearly all known biological processes and a range of pathological situations, which includes neuropathic pain. In chronic inflammatory pain scenarios, the involvement of sensory neurons, particularly thermoreceptors, and mechanoreceptors, plays a significant role in the development of neuropathic pain. This encompasses various conditions such as complex regional pain syndrome (CRPS), polyneuropathies, postherpetic neuralgia (PHN), and fibromyalgia syndrome. 63 Many miRNAs are now recognized as crucial regulators in the pathophysiology of this condition, controlling various aspects of neuroinflammation and neuronal gene expression. Researchers have directly observed distinct miRNA expression in the dorsal root ganglion (DRG) following the onset of neuropathic and inflammatory pain. 64 Hence, it is crucial to investigate the role of miRNAs in regulating the migration of immune cells and the process of neuroinflammation following nerve injury (Figures 1–3).
Figure 2.
■■■
Figure 1.
■■■
Figure 3.
■■■
Immune cell infiltration
Neuropathic pain is commonly linked to an immediate infiltration of immune cells following damage to peripheral nerves. Recent research indicates that miRNAs released by immune and nonimmune cells play a crucial role in immune regulation. In diabetic peripheral neuropathic pain, miR-590-3p has been found to control the entry of immune cells into neural tissues. 34 Similarly, after spinal nerve ligation (SNL), rats exhibited reduced levels of miR-214-3p in spinal astrocytes, leading to their hyperactivity due to the upregulation of CSF1. 32 Additionally, alterations in miRNA patterns in microglial cells suggest that these cells exhibit unique functions depending on particular tissue or level of the disease process.
Neuroinflammation
Preliminary studies have unveiled the dual capacity of miRNAs to activate or inhibit the immune system, suggesting their potential involvement in the progression and development of autoimmune and inflammatory conditions, including neuropathic pain. Throughout the various stages of these conditions, specific miRNAs have been observed to undergo alterations in expression. To exert their influence in neuropathic pain, miRNAs signal through multiple pathways, including TXNIP/NLRP3 inflammasome, MAP kinase, IRAK/TRAF6, TLR4/NF-κB, and TLR5 and TNF-α signaling. 65 Among the key receptors in the innate immune system, TLR plays a critical role by inducing the release of pro-inflammatory cytokines, initiating the production of inflammatory mediators responsible for pain, fever, and other inflammatory reactions, and exerting regulatory influence over the inflammatory cascade. 66 In cultured astrocytes from mice after spinal nerve ligation (SNL), exposure to IL-1β or TNF-α increased by Tumor necrosis factor receptor-associated factor 6 (TRAF6) levels. TRAF6 is a crucial mediator involved in TLR signaling, NF-κB activation, and the expression of proinflammatory cytokines and interferons. IL-1 receptor-associated kinase (IRAK) plays a role in MyD88 signaling. The interaction between TRAF6 and IRAK leads to further activation of Janus kinase N-terminal region (JAK) and nuclear factor κ-activated B-cell light chain enhancer (NF-κB). 67 By targeting miRNA-146a-5p, which inhibits specific sections of TRAF6 and IRAK1 mRNA 3′UTR in TLR signaling pathways and reduces their NF-κB activation, protein production, the NLRP3 inflammasome signaling pathway, and neuropathic pain levels are adversely affected. 68 By upregulating miR-381, neuropathic pain behaviors were relieved in rats with chronic constriction nerve injury (CCI) due to the inhibition of HMGB1 expression. Conversely, this positive effect was reversed when miR-381 inhibitors were utilized. 69 Similarly, overexpressing miR-362-3p led to significantly suppressing of proinflammatory cytokine levels by regulating BAMBI expression, thereby impeding the neuroinflammatory process and alleviating neuropathic pain in CCI mice. 70 The TGF-β family includes a group of cytokine secreted and have a crucial impact on multiple biological processes, including cell division, survival, differentiation, and migration. 71 Among the members of this family is transforming growth factor α (TNF-α), which is highly associated with neuropathic pain. Furthermore, a member of the C-C chemokine family, C-C motif chemokine ligand 2 (CCL2), exhibits a high affinity for C-C chemokine receptor 2 (CCR2). Recent research indicates that miR-183 overexpression decreases the CCLR2/TGF-α/CCL2 signaling axis, consequently suppressing the expression of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) as well as pain-related markers (Nav1.7, TRPV1, Nav1.8, Nav1.3) in the dorsal root ganglion (DRG). 29 In addition, a transcription factor known as zinc finger E-box-binding homeobox 1 (ZEB1) participates in diverse disease processes by repressing ZEB1 expression. A group of miRNAs, namely miR-136, miR-128-3p, miR-28-5p, miR-200b, miR-150, and miR-429, collaboratively regulate the advancement of neuropathic pain and neuroinflammation through the inhibition of ZEB1 expression.30,31 The G-protein-coupled receptor group, which includes CXCR4, plays a significant role in regulating the maturation of glial cells and neurons in the central nervous system. 72 The role of CXCR4 in various processes involved in sensing pain stimuli is becoming more apparent. Additionally, a versatile protein called thioredoxin-interacting protein (TXNIP) is critical in numerous cellular activities, encompassing metabolism, growth, division, and apoptosis. 73 The process of inflammation is initiated by the activation of a complex known as the NOD-like receptor family pyrin domain-containing protein 3 (NLRP3). This complex leads to the generation and release of pro-inflammatory cytokines, such as interleukin-1 (IL-1) and IL-18, playing a crucial role in the onset of inflammation. 74 MiR-23a exerts a significant impact on neuropathic pain (NP) by regulating spinal glial cells through the specific targeting of TXNIP/NLRP3 and CXCR4 on the inflammasome axis. Intrathecal administration of miR-23a effectively reduced hyperalgesia by mimicking the suppression of spinal CXCR4 through lentiviral delivery and inhibiting the overexpression of NLRP3 or TXNIP. 18 In a rat model of NP, the expression of miR-144 and miR-140 was found to be decreased in the dorsal root ganglion (DRG). Additionally, intrathecal injection of miR-144 and miR-140 agomir, which target RASP21 protein activator 1 (RASA1) and sphingosine-1-phosphate receptor 1 (S1PR1), respectively, led to reduced secretion of inflammatory factors and amelioration of hyperalgesia. 21 Moreover, in rats experiencing neuropathic pain following chronic nerve injury (CCI), miR-216a-5p alleviated the condition by targeting KDM3A and deactivating the Wnt/β-catenin signaling pathway. 75 In the context of CCI rats, miR-93 directly targets the 3′UTR of the Signal transducer and activator of transcription 3 (STAT3), thus preventing the progression of the disease. 27 NF-κB plays a vital role as a mediator in the inflammatory cascade. When activated, the NF-κB pathway triggers both M1 and M2 macrophages to release pro-inflammatory cytokines, thereby intensifying the overall inflammatory response. 76 On the other hand, MAPK, a mitogen-activated kinase, plays a significant role in various physiological and pathological processes, including neuropathic pain, through the phosphorylation of serine/threonine and tyrosine residues. 77 At the same time, a distinct set of proteins called cytokine signaling inhibitors (SOCS) maintains the equilibrium of Th2-Th1 cells, moderates the negative feedback of cytokine signaling, and lessens Th2-induced inflammation. SOCS1 and SOCS3 activation by various inflammatory and anti-inflammatory cytokines inhibits cytokine actions. 78 Several researchers have observed that inhibiting miR-221 or miR-155 alleviates neuroinflammation and neuropathic pain by increasing the expression of suppressor of cytokine signaling 1 (SOCS1) through inhibition of p38-MAPK and NF-κB. 79 Furthermore, in diabetic peripheral neuropathy (DPN), the inhibition of miR-221 led to pain reduction and a decrease in the expression of inflammatory factors (IL-1β, IL-6, PEG2, TNF-α, and BK) by specifically targeting SOCS3. 26 Similarly, in the context of oxaliplatin-induced peripheral neuropathic pain, miR-155 levels in the spinal cord rose. Interestingly, administering a miR-155 inhibitor through intrathecal injection led to a reduction in hyperalgesia in rats, potentially achieved by obstructing the oxidative stress-TRPA1 pathways. 28 Suppression or inhibition of miRNA-22 led to diminished mechanical allodynia and heat hyperalgesia triggered by Complete Freund’s adjuvant (CFA). On the contrary, the upregulation of miRNA-22 induced pain-related behaviors. The increased miRNA-22 directly interacted with the Mtf1 promoter, resulting in the activation of RNA polymerase II and an elevation in Mtf1 expression. The heightened expression of Mtf1 contributed to spinal central sensitization, as indicated by elevated levels of p-ERK1/2, GFAP, and c-Fos in the dorsal horn. 20 The results indicate that utilizing epigenetic approaches to modulate miRNAs for reducing neuroinflammation could present a hopeful and innovative therapeutic strategy for managing nociceptive hypersensitivity and neuropathic pain arising from peripheral nerve injuries.
Regulation of nerve regeneration
Following nerve injury, the preservation of neurons is a crucial prerequisite for the regeneration of nerves and the restoration of function. Prior studies have indicated that prolonged pain can result in damage or potential cell death of the spinal cord and peripheral nerve cell bodies. 80 Recent research has revealed that injured peripheral neurons can trigger the release of intrinsic neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and nerve growth factor (NGF), which facilitate neuronal survival and axon regeneration. 81 In this context, miRNAs also have a substantial impact on the process of nerve regeneration. Intriguingly, abnormal expression of several miRNAs that target genes crucial for nerve regeneration has been noted in the dorsal root ganglion (DRG). 82 Furthermore, it has been proposed that reducing miR-192-5p expression can increase the levels of the X-linked inhibitor of apoptosis protein (XIAP), thereby reducing nerve cell apoptosis and promoting regeneration after sciatic nerve injury (SNI). 37 Furthermore, the upregulation of miR-210 promotes the survival of neurons by inhibiting apoptosis through its interaction with ephrin-A3 (EFNA3), thus facilitating the process of axon regeneration. 39 MiR-135a and miR-135b effectively downregulate Kruppel-like factor 4 (KLF4), a potent inhibitor of axonal regeneration and outgrowth, thus promoting axonal outgrowth and cortical neuron migration. 35 In rats experiencing spinal cord injury (SCI), the simultaneous use of 3D fiber hydrogel scaffolds and continuous administration of a blend of axon miRNAs (miR-222, miR-431, and miR-132) led to notable enhancement in axon regeneration. 49 Prior research has demonstrated that elevated levels of miR-125b facilitate axon regeneration after spinal cord injury through regulation of the JAK/STAT pathway. Furthermore, miR-125b offers neuroprotection by diminishing neuronal apoptosis and dampening inflammatory responses. 38 In vivo, the deletion of miR-155 enhances injury-induced expression of SPRR1A, a gene related to regeneration, in neurons, and also reduces inflammatory signaling in macrophages, leading to improved axon regeneration. 36 Mice subjected to optic nerve compression exhibited notable augmentation of miR-19a levels within retinal ganglion cells, leading to a substantial enhancement of axon regeneration in vivo. 48 Furthermore, alterations in the expression of miR-199a-3p and miR-21, induced by injury, impact axon development by regulating protein synthesis systemically and within axons through the modulation of the mTOR/PTEN pathway. 42 The mTOR/PTEN pathway is a critical regulator of axonal regeneration. PTEN, a tumor suppressor phosphatase, functions as a PIP3 phosphatase, effectively restraining mTOR signaling. Meanwhile, mTOR, a serine/threonine protein kinase, facilitates protein synthesis and mRNA translation, leading to improved axon growth. 83 The combined action of these two elements collaboratively governs the growth of axons. MiRNA-21 regulates the expression of target genes, including EPHA4, TGF-I, and TIMP3, critical for promoting Schwann cell (SC) proliferation and facilitating axon regeneration in repairing injured peripheral nerves. 43 Moreover, the naturally occurring miR-26a within sensory neurons plays a role in aiding the regeneration of sensory axons following spinal cord injury (SNI) by facilitating the activation of Smad1 while inhibiting the expression of glycogen synthase kinase 3β (GSK3β). 45 On the other hand, numerous miRNAs act as negative regulators of neuronal regeneration, thus supporting the progression of neuropathic conditions. As an example, miR-21 regulates the epidermal growth factor receptor (EGFR) pathway, resulting in the excessive activation of astrocytes and the formation of glial scar tissue, which acts for obstacle to axon regeneration. 41 Blocking miR-455-5p leads to the inhibition of axonal growth and regeneration in sensory neurons of mice, and at the same time, reduces the activation of the GSK3β/Tau protein pathway. 47 miR-9 and miRlet-7 also hinder axonal regeneration by targeting the protein levels of Dcc and Ntn1, respectively. 44 Additionally, miR-9 is involved in regulating FoxP1 triggered by injury and has a role in axon regeneration. However, sensory neurons with high levels of endogenous miR-9 were found unable to regenerate their axons. 46 Additionally, a newly discovered mechanism of controlling the inherent potential for axon regeneration includes a reciprocal negative regulatory loop between SIRT1 and miR-138. 40 These discoveries introduce a new perspective for future investigations into axon regeneration in the context of neuropathic pain. Despite substantial advancements in comprehending the basic processes of peripheral nerve regeneration and leveraging these pathways for promoting regeneration following peripheral nerve injury (PNI), various challenges remain in devising therapies that facilitate comprehensive regeneration and functional restoration of neurons.
Regulation of neuronal ion channels
Following nerve fiber injury, alterations in the nerve endings and dorsal root ganglion (DRG) of the spinal cord may occur, affecting the structure and function of ion channels. These changes can lead to the abnormal discharge of neurons and subsequently result in pain. 84 Consequently, ion channels play a pivotal role in determining neuronal excitability, and they might be subject to miRNA regulation under painful conditions. It is worth noting that voltage-gated channels, implicated in the pain pathway, have become the primary focus of interventions for treating neuropathic pain.
Calcium channels
In addition to voltage-gated potassium and sodium channels, calcium channels are equally essential in the pain sensitization process after nerve injury, as they participate in neurotransmitter release, regulate neuronal excitability, and influence intracellular alterations, including gene activation. 85 In murine spinal neurons, the expression of miR-124a and miR-219, which negatively regulate spinal CaMKIIγ and the proinflammatory marker MeCP2, was significantly reduced after the development of inflammatory pain induced by either formalin or CFA injection. 53 Furthermore, a decrease in miR-103 expression was observed in the spinal neurons of SNL rats. This concomitantly impacted the translational levels of the three constituents forming the Cav1.2-L-type calcium channel, an ion channel linked to pain sensitization. 86 Moreover, miR-32-5p, through histone methylation-mediated targeting of Cav3.2 channels, downregulates its expression in trigeminal ganglion (TG) neurons, thereby controlling trigeminal neuropathic pain. 55 In mice, the miR-183 cluster exerts control over more than 80% of genes associated with neuropathic pain by modulating the auxiliary voltage-gated calcium channel subunits α2δ-1 and α2δ-2. This regulation affects basal mechanical sensitivity and mechanical allodynia. 54 These discoveries underscore the crucial involvement of miRNA-related channel abnormalities in developing nerve injury-induced neuropathic pain. They emphasize the significance of abnormal sensory input in maintaining neuropathic pain and suggest the potential efficacy of targeted chemo genetic silencing as a therapeutic approach for neuropathic pain treatment.
Voltage-gated potassium channels
Neurons’ excitability is significantly controlled by voltage-gated potassium channels, which govern action potential generation, neuronal firing frequency, and the release of neurotransmitters. 87 The miRNA cluster miR-17-92, comprising six distinct members, downregulates the expression of potassium channels, particularly type A currents, leading to decreased outward potassium currents and the development of mechanical allodynia. 88 Likewise, suppressing miR-137 leads to a decrease in mechanical allodynia and thermal hyperalgesia, normalizes Kv currents, reduces excessive neuronal activity in the dorsal root ganglion (DRG), and restores the expression of the potassium channel Kv1.2. 89 Furthermore, miR-183-5p plays a role in controlling CCI-induced neuropathic pain by suppressing the expression of TREK-1, a Kv channel. 50
Voltage-gated sodium channels
The Nav1.3 voltage-gated sodium channel, a variant susceptible to tetrodotoxin encoded by SCN3A, produces fast-repriming sodium ion currents. These currents can promote repetitive firing patterns and abnormal discharges in injured neurons, leading to heightened neuronal excitability and closely linked to neuropathic pain. 90 Similar sodium-ion channels include Nav1.6, encoded by SCN8A, and Nav1.7, encoded by SCN9A. 91 MiR-96 administration via intrathecal delivery suppresses the upregulation of Nav1.3 caused by CCI. Additionally, miR-96 has been found to decrease the expression of Nav1.3 mRNA in embryonic DRG neurons in in vitro studies. 61 Additionally, miR-384-5p regulates the emergence of neuropathic pain by controlling SCN3A. 59 In rats, miR-182 effectively reduced the neuropathic pain induced by SNI by regulating Nav1.7. 60 Remarkably, the heightened presence of miR-30b resulted in a marked decline in the levels of Nav1.7, Nav1.6, and Nav1.3 in both dorsal root ganglion (DRG) neurons and the spinal cord, thereby leading to a considerable alleviation of neuropathic pain induced by spinal nerve ligation (SNL) or oxaliplatin. 57 Similarly, the heightened presence of miR-183 reduced osteoarthritic pain by suppressing the synthesis of Nav1.8, Nav1.3, and Nav1.7. 29 Moreover, elevated levels of miR-7a in primary sensory neurons of damaged DRGs hindered the increase in the β2 subunit of voltage-gated sodium channels. They reverted the persistent hyperexcitability of pain-sensing neurons to their regular state. 51
Role of exosomal miRNAs in neuropathic pain
Exosomes (Exos) are a group of nanosized extracellular vesicles (EVs) ranging in size from 40 to 200 nm, which are released by various cell types and participate in paracrine interactions among different cell types, such as glial cells, mesenchymal stem cells, neurons, leukocytes and endothelial cells. 92 Functioning as unique secreted agents, these biological nanoparticles are abundant in various genetic components like miRNAs, long noncoding RNA, lipids, and proteins. They can easily spread throughout biofluids, impacting biochemical reactions and cell survival in normal and disease-related circumstances, particularly in neurodegenerative and inflammatory conditions. 93 Notably, exosomal miRNAs can be found in significant quantities in various bodily fluids like blood, saliva, breast milk, and urine. 94 Pain often arises from inflammation, with various cytokines, chemokines, and other factors contributing to the onset of acute inflammatory pain. In cases of chronic inflammation, the processes leading to peripheral and cerebral sensitization can be initiated. 95 Exosomal miRNAs, once produced, can be transported to different sites and act on neurons, microglia, macrophages, or other tissue cells. These entities control neuropathic pain through their impact on the release of inflammatory substances and oxidative stress, as well as their ability to regulate neural remodeling and nerve regeneration processes. Exosomes can modulate the release of nociceptive mediators from cells, which are known to be involved in neuroinflammation and are recognized for their role in sensitizing sensory terminals. 96 Immunological cells, such as T lymphocytes and antigen-presenting dendritic cells (DCs), are capable of releasing and absorbing exosomal miRNAs, suggesting that exosomal miRNA transfer could serve as a novel mode of intercellular communication. 97 Consequently, exosomal miRNA transmission holds significant implications for various systems and processes, including immune responses and neuron-glia communication. 98 Fascinatingly, these exosomal miRNAs possess the ability to elicit both anti-inflammatory and proinflammatory responses. As previously discussed, specific exosomal miRNAs can release cytokines or other proinflammatory agents, influencing target organs directly. 99 For instance, exosomes derived from neutrophils and synovial fibroblasts, and chondrocytes have been observed to stimulate macrophages to produce IL-1 and metalloproteinases. Additionally, the production of these inflammatory substances has been linked to the transfer of miR-449a-5p and miR-206 within exosomes’ contents. 100 MiR-449a-5p plays a crucial role in inhibiting ATG4B, which regulates macrophage autophagy, triggers inflammasome activation, and exacerbates the inflammatory response. Studies have indicated that serum exosomes from neuropathic mice exhibit increased levels of miR-21. Further investigations revealed macrophage-derived exosomes containing miR-21-5p promote pyroptosis through A20, leading to a proinflammatory phenotype and aggravating podocyte damage in diabetic nephropathy mice. 101 Notably, intrathecal addition of miR-21-5p antagomir or conditional deletion of miR-21 in sensory neurons reduced hyperalgesia and macrophage recruitment in the DRG. A20 acts as an inhibitor of the NF-κB signaling pathway. Similarly, research showed that DRG sensory neurons release miR-23a-enriched extracellular vesicles (EVs) upon nerve damage, which are then taken up by macrophages to promote M1 polarization in vitro. Moreover, intrathecal delivery of an EV-miR-23a antagomir, which blocks A20 to enhance NF-κB signaling, reduced M1 macrophages, and improved neuropathic hyperalgesia. 102 Conversely, specific exosomal miRNAs exhibit anti-inflammatory and pain-relieving properties in chronic pain models by transporting healing substances to damaged neurons in both the central nervous system (CNS) and peripheral nervous system (PNS). 103 The beginning of inflammation is thought to be the main cause of pain. Furthermore, exosomal miRNAs can suppress the generation of proinflammatory cytokines like PGE2, IL-1β, TNF-α and IL-6 at the injury site, while stimulating the release of IL-10, leading to pain relief effects. 93 These miRNAs possess the ability to modulate nociception, and when administered intrathecally, miR-25, miR-103, miR-544, miR-124, and miR-23b have been shown to reduce inflammatory and neuropathic pain by modifying the intracellular activities of neurons, astrocytes, and microglia.104,105 MiR-124 suppresses GRK2 expression, thus controlling the phenotypic balance between M1 and M2 cells in the spinal cord. Similarly, akin to certain neurotrophic factors like BDNF, GDNF, NGF, FGF-1, and IGF-1, exosomal miRNAs possess the capacity to enhance axonal growth and neuronal viability, intensifying therapeutic outcomes. 106 Until now, the targeted and modular EV loading (TAMEL) approach has not been utilized in experimental pain research. Despite numerous studies attempting to shed light on the function of exosomal miRNAs in neuropathic pain, much remains to be unraveled about their cellular and molecular roles, as well as their downstream targets.
The emerging role of exosomal RNAs in neuropathic pain
Common pain relievers like acetaminophen, nonsteroidal anti-inflammatory drugs (NSAIDs), local anesthetics, and opioids can effectively manage acute pain. However, when it comes to neuropathic pain, these options often fall short due to limited efficacy or unwanted side effects. 107 Therefore, current research endeavors should prioritize the discovery and development of mechanism-based therapies aimed at alleviating neuropathic pain. Laboratory investigations reveal that exosomes can regulate neuropathic pain and can be released by various cell types, including stem cells. 103 Moreover, evidence indicates that exosomes derived from stem cells can closely replicate the functional effects of their parent cells, making them vital players in tissue repair processes. 108 Exosomes derived from stem cells offer a novel therapeutic approach and research focus for neuropathic pain, as they not only aid in nerve repair but also eliminate the risks associated with stem cell transplantation, such as immunosuppression, genetic alterations, and malignant transformation, thanks to their paracrine effects. 109 These stem cell-derived exosomes are believed to transfer beneficial neurotrophic factors like IGF-1, FGF-1, NGF, BDNF, and GDNF to injured neurons. Intrathecal administration of mesenchymal stem cell exosomes in rats with spinal cord injuries has demonstrated a reduction in neuropathic pain by shifting microglia from the M1 to the M2 phenotype and inhibiting the release of inflammatory cytokines like NF-κB, IL-6, IL-1, and TNF-α. 110 The TLR4/miR-216a-5p axis has been shown to have a regulatory impact on microglial polarization. 37 Mesenchymal stem cells (MSCs) are an up-and-coming stem cell type for treating various ischemic conditions and tissue damage due to their versatile differentiation potential and strong immune regulatory functions. 111 Recent research in the laboratory revealed that MSCs can migrate to injured nerve tissues and promote the regeneration of damaged neurons. 112 Exosomes derived from MSCs regulate the growth of neurites by affecting both the quantity and overall length of neurites through the transmission of miR-133b to nerve cells.113,114 Moreover, exosomes containing an abundance of miR-17-92 clusters have the potential to boost neuroplasticity and improve functional recovery by targeting PTEN, thereby activating the GSK-3β/Akt/PI3K/mTOR signaling pathway. 115 Likewise, small exosomes derived from Schwann cells (SCs) that contain miR-21-5p exert a negative regulatory effect on PTEN, leading to improved growth and survival of sensory neurons. 116 In order to promote neurite outgrowth in laboratory settings and support nerve regeneration in living organisms, secreted extracellular vehicles (EVs) from SCs carry miR-23b-3p from mechanically stimulated SCs to neurons, leading to a decrease in the levels of neuronal neuropilin 1 (Nrp1). 117 Exosomes derived from umbilical cord mesenchymal stem cells enhance axon regrowth and functional improvement in the spinal cord of rats by targeting Cblb/Cbl-mediated NGF/TrkA signaling through miR-199a-3p/145-5p. 118 After spinal cord injury, neural stem cell-released exosomes, stimulated by IGF-1, reduce apoptosis and promote nerve regeneration, partially through a YY1/miR-219a-2-3p mechanism. 119 Likewise, miR-181c-5p acts as a negative regulator of Bcl-2-interacting cell death mediators (BIM), thereby preventing neuronal apoptosis and maintaining cortical neuron health, which ultimately facilitating axon regeneration. 120 A recent investigation revealed that stem cell-derived exosomes enriched with miR-29 could alleviate proinflammatory responses in a rat model of osteoarthritis. 121 Similarly, miR-199-3p overexpression in mice attenuated postherpetic neuralgia induced by TRX by targeting MECP2. 122 Microglial cells incorporated exomiR-181c-5p, which prevented the release of inflammatory substances. Intrathecal treatment with exomiR-181c-5p in CCI rats reduced neuroinflammatory symptoms and neuropathic pain. 70 To counter neuroinflammation, exosomes originating from human umbilical cord MSCs increased the expression of autophagy-related proteins (LC3-II and beclin1) and hindered NLRP3 inflammasome activation via the TRAF6/miR-146a-5p pathway in the spinal cord dorsal horn. 123 Additionally, xenogenic injection of human MSC exosomes enriched with miR-26a-5p led to the downregulation of cyclooxygenase-2 (PTGS2) in rat synovial fibroblasts, resulting in reduced pathogenic alterations. 124 Exosomes derived from MSCs exhibit pain-relieving properties in chronic pain models by delivering particular miRNAs to both the central nervous system (CNS) and peripheral nervous system (PNS). 125 Exosomal miRNAs offer distinct advantages as biomarkers compared to freely circulating miRNAs: firstly, exosomes harbor a wide variety of miRNAs, making them reliable vehicles for miRNA research 1 ; secondly, the bilayer membrane structure of exosomes enhances miRNA stability, susceptibility to amplification, and reduces unfavorable outcomes 2 ; and lastly, exosomes can efficiently traverse the blood-brain or blood-spinal cord barriers. 4 Despite the potential benefits of MSC-derived exosomes, the primary challenge in stem cell therapy remains the limited therapeutic effectiveness due to the low survival rate of transplanted cells in injured tissues. The results indicate that stem cell-derived exosomal miRNAs can control pain by diminishing proinflammatory cytokines and fostering neuronal regeneration and differentiation, presenting a fresh and innovative therapeutic strategy for the treatment of nerve injuries. In conclusion, neuropathic pain poses a major risk to patients’ physical and emotional well-being, and the absence of reliable and efficient treatment methods adds to the difficulty of addressing this therapeutic challenge. The extensive investigation of miRNA in the development of neuropathic pain has opened promising avenues for clinical translation, particularly due to its remarkable impact on neuroinflammation, nerve regeneration, and other aspects, offering hope for improved clinical management of neuropathic pain. However, challenges such as miRNA’s bioactivity, stability, safety, and tissue specificity must be addressed before its widespread therapeutic application. Efficient transmembrane delivery and careful consideration of enzymatic reactions are additional critical factors during miRNA targeting. As a result, extensive focus is currently directed toward investigating delivery agents, such as exosomes, liposomes, viral vectors, miRNA mimics, and inhibitors. Patient safety remains a top priority during clinical applications, necessitating a thorough assessment of the potential immunological response triggered by exogenous miRNA treatment. Additional preclinical investigations and clinical trials are crucial to support the successful application of these findings in clinical settings, as the current research is primarily confined to studies conducted in cell cultures and animal models. With ongoing research into miRNA mechanisms and the utilization of advanced technologies like miRNA gene chips and high-throughput methods, miRNAs can become novel biological markers for disease diagnosis and offer new targets and strategies for understanding and treating neuropathic pain in the future.
Footnotes
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD
Amir Azimian https://orcid.org/0000-0002-2532-8198
References
- 1.Scholz J, Finnerup NB, Attal N, Aziz Q, Baron R, Bennett MI, Benoliel R, Cohen M, Cruccu G, Davis KD, Evers S, First M, Giamberardino MA, Hansson P, Kaasa S, Korwisi B, Kosek E, Lavand'homme P, Nicholas M, Nurmikko T, Perrot S, Raja SN, Rice ASC, Rowbotham MC, Schug S, Simpson DM, Smith BH, Svensson P, Vlaeyen JWS, Wang SJ, Barke A, Rief W, Treede RD, Classification Committee of the Neuropathic Pain Special Interest Group NeuPSIG . The IASP classification of chronic pain for ICD-11: chronic neuropathic pain. Pain 2019; 160(1): 53–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Yi M-H, Liu YU, Liu K, Chen T, Bosco DB, Zheng J, Xie M, Zhou L, Qu W, Wu LJ. Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Brain Behav Immun 2021; 92: 78–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Walters ET. Neuroinflammatory contributions to pain after SCI: roles for central glial mechanisms and nociceptor-mediated host defense. Exp Neurol 2014; 258: 48–61. [DOI] [PubMed] [Google Scholar]
- 4.Huh Y, Ji R-R, Chen G. Neuroinflammation, bone marrow stem cells, and chronic pain. Front Immunol 2017; 8: 1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Li J, Wei G-H, Huang H, Lan Y-P, Liu B, Liu H, Zhang W, Zuo YX. Nerve injury-related autoimmunity activation leads to chronic inflammation and chronic neuropathic pain. Anesthesiology 2013; 118(2): 416. [DOI] [PubMed] [Google Scholar]
- 6.Bernardo BC, Ooi JY, Lin RC, McMullen JR. miRNA therapeutics: a new class of drugs with potential therapeutic applications in the heart. Future Med Chem 2015; 7(13): 1771. [DOI] [PubMed] [Google Scholar]
- 7.Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 2017; 16(3): 203. [DOI] [PubMed] [Google Scholar]
- 8.Li Y, Yin Z, Fan J, Zhang S, Yang W. The roles of exosomal miRNAs and lncRNAs in lung diseases. Signal Transduct Target Ther 2019; 4(1): 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hill M, Tran N. miRNA interplay: mechanisms and consequences in cancer. Dis Model Mech 2021; 14(4): dmm047662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Olive V, Minella AC, He L. Outside the coding genome, mammalian microRNAs confer structural and functional complexity. Sci Signal 2015; 8(368): re2, re2-re. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Saliminejad K, Khorram Khorshid HR, Soleymani Fard S, Ghaffari SH. An overview of microRNAs: biology, functions, therapeutics, and analysis methods. J Cell Physiol 2019; 234(5): 5451. [DOI] [PubMed] [Google Scholar]
- 12.Lutz BM, Bekker A, Tao Y-X. Noncoding RNAs: new players in chronic pain. Anesthesiology 2014; 121(2): 409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Li Y, Huang J, Guo M, Zuo X. MicroRNAs regulating signaling pathways: potential biomarkers in systemic sclerosis. Dev Reprod Biol 2015; 13(4): 234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lima RT, Busacca S, Almeida GM, Gaudino G, Fennell DA, Vasconcelos MH. MicroRNA regulation of core apoptosis pathways in cancer. Eur J Cancer 2011; 47(2): 163. [DOI] [PubMed] [Google Scholar]
- 15.Barwari T, Joshi A, Mayr M. MicroRNAs in cardiovascular disease. J Am Coll Cardiol 2016; 68(23): 2577. [DOI] [PubMed] [Google Scholar]
- 16.Wang X, Zhou Y, Gao Q, Ping D, Wang Y, Wu W, Lin X, Fang Y, Zhang J, Shao A. The role of exosomal microRNAs and oxidative stress in neurodegenerative diseases. Oxid Med Cell Longev 2020; 2020: 3232869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhang X, Guo H, Xie A, Liao O, Ju F, Zhou Y. MicroRNA-144 relieves chronic constriction injury-induced neuropathic pain via targeting RASA1. Biotechnol Appl Biochem 2020; 67(2): 294–302. [DOI] [PubMed] [Google Scholar]
- 18.Pan Z, Shan Q, Gu P, Wang XM, Tai LW, Sun M, Luo X, Sun L, Cheung CW. miRNA-23a/CXCR4 regulates neuropathic pain via directly targeting TXNIP/NLRP3 inflammasome axis. J Neuroinflammation 2018; 15: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Xia L, Zhang Y, Dong T. Inhibition of microRNA-221 alleviates neuropathic pain through targeting suppressor of cytokine signaling 1. J Mol Neurosci 2016; 59: 411. [DOI] [PubMed] [Google Scholar]
- 20.Hao L-Y, Zhang M, Tao Y, Xu H, Liu Q, Yang K, Wei R, Zhou H, Jin T, Liu X-D, Xue Z, Shen W, Cao J-L, Pan Z. miRNA-22 upregulates Mtf1 in dorsal horn neurons and is essential for inflammatory pain. Oxid Med Cell Longev 2022; 2022: 8622388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li J, Zhu Y, Ma Z, Liu Y, Sun Z, Wu Y. miR-140 ameliorates neuropathic pain in CCI rats by targeting S1PR1. J Recept Signal Transduct Res 2021; 41(4): 401. [DOI] [PubMed] [Google Scholar]
- 22.Yan XT, Lu JM, Wang Y, Cheng XL, He XH, Zheng WZ, Chen H, Wang YL. XIST accelerates neuropathic pain progression through regulation of miR-150 and ZEB1 in CCI rat models. J Cell Physiol 2018; 233(8): 6098. [DOI] [PubMed] [Google Scholar]
- 23.Yan XT, Zhao Y, Cheng XL, He XH, Wang Y, Zheng WZ, Chen H, Wang YL. Inhibition of miR-200b/miR-429 contributes to neuropathic pain development through targeting zinc finger E box binding protein-1. J Cell Physiol 2018; 233(6): 4815. [DOI] [PubMed] [Google Scholar]
- 24.Tan Y, Yang J, Xiang K, Tan Q, Guo Q. Suppression of microRNA-155 attenuates neuropathic pain by regulating SOCS1 signalling pathway. Neurochem Res 2015; 40: 550. [DOI] [PubMed] [Google Scholar]
- 25.Zhang W, Zhou L, Zhang C. LncRNA Miat promotes neuropathic pain through miR-362-3p/BAMBI signaling axis. Exp Cell Res 2022; 420(2): 113359. [DOI] [PubMed] [Google Scholar]
- 26.Wu X, Wang X, Yin Y, Zhu L, Zhang F, Yang J. Investigation of the role of miR-221 in diabetic peripheral neuropathy and related molecular mechanisms. Adv Clin Exp Med 2021; 30(6): 623. [DOI] [PubMed] [Google Scholar]
- 27.Yan X-T, Ji L-J, Wang Z, Wu X, Wang Q, Sun S, Lu JM, Zhang Y. MicroRNA-93 alleviates neuropathic pain through targeting signal transducer and activator of transcription 3. Int Immunopharmacol 2017; 46: 156. [DOI] [PubMed] [Google Scholar]
- 28.Miao F, Wang R, Cui G, Li X, Wang T, Li X. Engagement of microRNA-155 in exaggerated oxidative stress signal and TRPA1 in the dorsal horn of the spinal cord and neuropathic pain during chemotherapeutic oxaliplatin. Neurotox Res 2019; 36: 712. [DOI] [PubMed] [Google Scholar]
- 29.Tao Z, Zhou Y, Zeng B, Yang X, Su M. MicroRNA-183 attenuates osteoarthritic pain by inhibiting the TGFα-mediated CCL2/CCR2 signalling axis. Bone Joint Res 2021; 10(8): 548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bao Y, Wang S, Xie Y, Jin K, Bai Y, Shan S. MiR-28-5p relieves neuropathic pain by targeting Zeb1 in CCI rat models. J Cell Biochem 2018; 119(10): 8555. [DOI] [PubMed] [Google Scholar]
- 31.Shen F, Zheng H, Zhou L, Li W, Zhang Y, Xu X. LINC00657 expedites neuropathic pain development by modulating miR-136/ZEB1 axis in a rat model. J Cell Biochem 2019; 120(1): 1000. [DOI] [PubMed] [Google Scholar]
- 32.Liu L, Xu D, Wang T, Zhang Y, Yang X, Wang X, Tang Y. Epigenetic reduction of miR-214-3p upregulates astrocytic colony-stimulating factor-1 and contributes to neuropathic pain induced by nerve injury. Pain 2020; 161(1): 96–108. [DOI] [PubMed] [Google Scholar]
- 33.Wang Z, Liu F, Wei M, Qiu Y, Ma C, Shen L, Huang Y. Chronic constriction injury-induced microRNA-146a-5p alleviates neuropathic pain through suppression of IRAK1/TRAF6 signaling pathway. J Neuroinflammation 2018; 15(1): 179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wu Y, Gu Y, Shi B. miR-590-3p Alleviates diabetic peripheral neuropathic pain by targeting RAP1A and suppressing infiltration by the T cells. Acta Biochim Pol 2020; 67(4): 587. [DOI] [PubMed] [Google Scholar]
- 35.Van Battum EY, Verhagen MG, Vangoor VR, Fujita Y, Derijck AA, O'Duibhir E, Giuliani G, de Gunst T, Adolfs Y, Lelieveld D, Egan D, Schaapveld RQJ, Yamashita T, Pasterkamp RJ. An image-based miRNA screen identifies miRNA-135s as regulators of CNS axon growth and regeneration by targeting Krüppel-like factor 4. J Neurosci 2018; 38(3): 613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gaudet AD, Mandrekar-Colucci S, Hall JC, Sweet DR, Schmitt PJ, Xu X, Guan Z, Mo X, Guerau-de-Arellano M, Popovich PG. miR-155 deletion in mice overcomes neuron-intrinsic and neuron-extrinsic barriers to spinal cord repair. J Neurosci 2016; 36(32): 8516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liu X, Cui X, Guan G, Dong Y, Zhang Z. microRNA-192-5p is involved in nerve repair in rats with peripheral nerve injury by regulating XIAP. Cell Cycle 2020; 19(3): 326. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 38.Dai J, Xu L-J, Han G-D, Sun H-L, Zhu G-T, Jiang H-T, Yu GY, Tang XM. MicroRNA-125b promotes the regeneration and repair of spinal cord injury through regulation of JAK/STAT pathway. Eur Rev Med Pharmacol Sci 2018; 22(3): 582–589. [DOI] [PubMed] [Google Scholar]
- 39.Hu Y-W, Jiang J-J, Wang R-Y, Wang RY, Tu GJ. MicroRNA-210 promotes sensory axon regeneration of adult mice in vivo and in vitro. Neurosci Lett 2016; 622: 61. [DOI] [PubMed] [Google Scholar]
- 40.Liu C-M, Wang R-Y, Jiao Z-X, Jiao ZX, Zhang BY, Zhou FQ. MicroRNA-138 and SIRT1 form a mutual negative feedback loop to regulate mammalian axon regeneration. Genes Dev 2013; 27(13): 1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li H-J, Pan Y-B, Sun Z-L, Sun Y-Y, Yang X-T, Feng D-F. Inhibition of miR-21 ameliorates excessive astrocyte activation and promotes axon regeneration following optic nerve crush. Neuropharmacology 2018; 137: 33–49. [DOI] [PubMed] [Google Scholar]
- 42.Kar AN, Lee S-J, Sahoo PK, Thames E, Yoo S, Houle JD, Twiss JL. MicroRNAs 21 and 199a-3p regulate axon growth potential through modulation of Pten and mTor mRNAs. Eneuro 2021; 8(4): 155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ning X-J, Lu X-H, Luo J-C, Chen C, Gao Q, Li Z-Y, Wang H. Molecular mechanism of microRNA-21 promoting Schwann cell proliferation and axon regeneration during injured nerve repair. RNA Biol 2020; 17(10): 1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang X, Chen Q, Yi S, Liu Q, Zhang R, Wang P, Qian T, Li S. The microRNAs let-7 and miR-9 down-regulate the axon-guidance genes Ntn1 and Dcc during peripheral nerve regeneration. J Biol Chem 2019; 294(10): 3489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Jiang J, Liu C, Zhang B, Wang X, Zhang M, Zhang S, Zhang SR, Hall P, Hu YW, Zhou FQ. MicroRNA-26a supports mammalian axon regeneration in vivo by suppressing GSK3β expression. Cell Death Dis 2015; 6(8): e1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jiang J, Hu Y, Zhang B, Shi Y, Zhang J, Wu X, Yao P. MicroRNA-9 regulates mammalian axon regeneration in peripheral nerve injury. Mol Pain 2017; 13: 1744806917711612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Su H, Xiaohui X, He X, Liu C, Wang G, Zhou C. The miR-455-5p/ERα36 axis regulates mammalian neuronal viability and axonal regeneration. Neurosci Lett 2020; 735: 135159. [DOI] [PubMed] [Google Scholar]
- 48.Mak HK, Yung JS, Weinreb RN, Ng SH, Cao X, Ho TY, Ng TK, Chu WK, Yung WH, Choy KW, Wang CC, Lee TL, Leung CKS. MicroRNA-19a-PTEN axis is involved in the developmental decline of axon regenerative capacity in retinal ganglion cells. Mol Ther Nucleic Acids 2020; 21: 251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang N, Lin J, Lin VPH, Milbreta U, Chin JS, Chew EGY, Lian MM, Foo JN, Zhang K, Wu W, Chew SY. A 3D fiber-hydrogel based non-viral gene delivery platform reveals that microRNAs promote axon regeneration and enhance functional recovery following spinal cord injury. Adv Sci 2021; 8(15): 2100805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shi D-N, Yuan Y-T, Ye D, Kang L-M, Wen J, Chen H-P. MiR-183-5p alleviates chronic constriction injury-induced neuropathic pain through inhibition of TREK-1. Neurochem Res 2018; 43: 1143. [DOI] [PubMed] [Google Scholar]
- 51.Sakai A, Saitow F, Miyake N, Miyake K, Shimada T, Suzuki H. miR-7a alleviates the maintenance of neuropathic pain through regulation of neuronal excitability. Brain 2013; 136(9): 2738. [DOI] [PubMed] [Google Scholar]
- 52.Li Z, Yang CS, Nakashima K, Rana TM. Small RNA-mediated regulation of iPS cell generation. The EMBO journal 2011; 30(5): 823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pan Z, Zhu L-J, Li Y-Q, Hao L-Y, Yin C, Yang J-X, Guo Y, Zhang S, Hua L, Xue ZY, Zhang H, Cao JL. Epigenetic modification of spinal miR-219 expression regulates chronic inflammation pain by targeting CaMKIIγ. J Neurosci 2014; 34(29): 9476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Peng C, Li L, Zhang M-D, Bengtsson Gonzales C, Parisien M, Belfer I, Usoskin D, Abdo H, Furlan A, Häring M, Lallemend F, Harkany T, Diatchenko L, Hökfelt T, Hjerling-Leffler J, Ernfors P. miR-183 cluster scales mechanical pain sensitivity by regulating basal and neuropathic pain genes. Science 2017; 356(6343): 1168. [DOI] [PubMed] [Google Scholar]
- 55.Qi R, Cao J, Sun Y, Li Y, Huang Z, Jiang D, Jiang XH, Snutch TP, Zhang Y, Tao J. Histone methylation-mediated microRNA-32-5p down-regulation in sensory neurons regulates pain behaviors via targeting Cav3. 2 channels. Proc Natl Acad Sci U S A 2022; 119(14): e2117209119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kynast KL, Russe OQ, Möser CV, Geisslinger G, Niederberger E. Modulation of central nervous system–specific microRNA-124a alters the inflammatory response in the formalin test in mice. Pain® 2013; 154(3): 368. [DOI] [PubMed] [Google Scholar]
- 57.Shao J, Cao J, Wang J, Ren X, Su S, Li M, Li Z, Zhao Q, Zang W. MicroRNA-30b regulates expression of the sodium channel Nav1. 7 in nerve injury-induced neuropathic pain in the rat. Mol Pain 2016; 12: 1744806916671523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Su S, Shao J, Zhao Q, Ren X, Cai W, Li L, Bai Q, Chen X, Xu B, Wang J, Cao J, Zang W. MiR-30b attenuates neuropathic pain by regulating voltage-gated sodium channel Nav1. 3 in rats. Front Mol Neurosci 2017; 10: 126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ye G, Zhang Y, Zhao J, Chen Y, Kong L, Sheng C, Yuan L. miR-384-5p ameliorates neuropathic pain by targeting SCN3A in a rat model of chronic constriction injury. Neurol Res 2020; 42(4): 299–307. [DOI] [PubMed] [Google Scholar]
- 60.Cai W, Zhao Q, Shao J, Zhang J, Li L, Ren X, Su S, Bai Q, Li M, Chen X, Wang J, Cao J, Zang W. MicroRNA-182 alleviates neuropathic pain by regulating Nav1. 7 following spared nerve injury in rats. Sci Rep 2018; 8(1): 16750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chen H-P, Zhou W, Kang L-M, Yan H, Zhang L, Xu B-H, Cai WH. Intrathecal miR-96 inhibits Nav1. 3 expression and alleviates neuropathic pain in rat following chronic construction injury. Neurochem Res 2014; 39: 76–83. [DOI] [PubMed] [Google Scholar]
- 62.Li L, Shao J, Wang J, Liu Y, Zhang Y, Zhang M, Zhang J, Ren X, Su S, Li Y, Cao J, Zang W. MiR-30b-5p attenuates oxaliplatin-induced peripheral neuropathic pain through the voltage-gated sodium channel Nav1. 6 in rats. Neuropharmacology 2019; 153: 111. [DOI] [PubMed] [Google Scholar]
- 63.Sayed D, Abdellatif M. MicroRNAs in development and disease. Physiol Rev 2011; 91(3): 827. [DOI] [PubMed] [Google Scholar]
- 64.Fang Z-H, Liao H-L, Tang Q-F, Liu Y-J, Zhang Y-Y, Lin J, Yu HP, Zhou C, Li CJ, Liu F, Shen JF. Interactions among non-coding RNAs and mRNAs in the trigeminal ganglion associated with neuropathic pain. J Pain Res 2022; 15: 2967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gada Y, Pandey A, Jadhav N, Ajgaonkar S, Mehta D, Nair S. New vistas in microRNA regulatory interactome in neuropathic pain. Front Pharmacol 2022; 12: 778014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tang X, Xu Q, Yang S, Huang X, Wang L, Huang F, Luo J, Zhou X, Wu A, Mei Q, Zhao C, Wu J. Toll-like receptors and thrombopoiesis. Int J Mol Sci 2023; 24(2): 1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zarezadeh Mehrabadi A, Aghamohamadi N, Khoshmirsafa M, Aghamajidi A, Pilehforoshha M, Massoumi R, Falak R. The roles of interleukin-1 receptor accessory protein in certain inflammatory conditions. Immunology 2022; 166(1): 38–46. [DOI] [PubMed] [Google Scholar]
- 68.Hou J, Deng Q, Deng X, Zhong W, Liu S, Zhong Z. MicroRNA-146a-5p alleviates lipopolysaccharide-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production via the regulation of TRAF6 and IRAK1 in human umbilical vein endothelial cells (HUVECs). Ann Transl Med 2021; 9(18): 1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Xia LX, Ke C, Lu JM. NEAT1 contributes to neuropathic pain development through targeting miR-381/HMGB1 axis in CCI rat models. J Cell Physiol 2018; 233(9): 7103. [DOI] [PubMed] [Google Scholar]
- 70.Zhang Y, Ye G, Zhao J, Chen Y, Kong L, Sheng C, Yuan L. Exosomes carried miR-181c-5p alleviates neuropathic pain in CCI rat models. An Acad Bras Cienc 2022; 94: e20210564. [DOI] [PubMed] [Google Scholar]
- 71.Nickel J, Ten Dijke P, Mueller TD. TGF-β family co-receptor function and signaling. Acta Biochim Biophys Sin 2018; 50(1): 12–36. [DOI] [PubMed] [Google Scholar]
- 72.Bianchi ME, Mezzapelle R. The chemokine receptor CXCR4 in cell proliferation and tissue regeneration. Front Immunol 2020; 11: 2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wondafrash DZ, Nire’a AT, Tafere GG, Desta DM, Berhe DA, Zewdie KA. Thioredoxin-interacting protein as a novel potential therapeutic target in diabetes mellitus and its underlying complications. Diabetes Metab Syndr Obes 2020; 13: 43–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci 2016; 41(12): 1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wang Y, Lai X, Wu D, Liu B, Wang N, Rong L. Umbilical mesenchymal stem cell-derived exosomes facilitate spinal cord functional recovery through the miR-199a-3p/145-5p-mediated NGF/TrkA signaling pathway in rats. Stem Cell Res Ther 2021; 12: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Poladian N, Orujyan D, Narinyan W, Oganyan AK, Navasardyan I, Velpuri P, Chorbajian A, Venketaraman V. Role of NF-κB during Mycobacterium tuberculosis infection. Int J Mol Sci 2023; 24(2): 1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang H, Zhou Y, Wen D, Wang J. Noncoding RNAs: master regulator of fibroblast to myofibroblast transition in fibrosis. Int J Mol Sci 2023; 24(2): 1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sobah ML, Liongue C, Ward AC. SOCS proteins in immunity, inflammatory diseases, and immune-related cancer. Front Med 2021; 8: 727987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liu W-z, Ma Z-j, Li J-r, Kang X-w. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther 2021; 12(1): 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cohen SP, Vase L, Hooten WM. Chronic pain: an update on burden, best practices, and new advances. Lancet 2021; 397(10289): 2082. [DOI] [PubMed] [Google Scholar]
- 81.Keefe KM, Sheikh IS, Smith GM. Targeting neurotrophins to specific populations of neurons: NGF, BDNF, and NT-3 and their relevance for treatment of spinal cord injury. Int J Mol Sci 2017; 18(3): 548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bai G, Ambalavanar R, Wei D, Dessem D. Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain. Mol Pain 2007; 3: 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tang BL. Promoting axonal regeneration through exosomes: an update of recent findings on exosomal PTEN and mTOR modifiers. Brain Res Bull 2018; 143: 123. [DOI] [PubMed] [Google Scholar]
- 84.Jiang M, Wang Y, Wang J, Feng S, Wang X. The etiological roles of miRNAs, lncRNAs, and circRNAs in neuropathic pain: a narrative review. J Clin Lab Anal 2022; 36(8): e24592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rahman W, Dickenson AH. Voltage gated sodium and calcium channel blockers for the treatment of chronic inflammatory pain. Neurosci Lett 2013; 557 Pt A: 19–26. [DOI] [PubMed] [Google Scholar]
- 86.Favereaux A, Thoumine O, Bouali-Benazzouz R, Roques V, Papon MA, Salam SA, Drutel G, Léger C, Calas A, Nagy F, Landry M. Bidirectional integrative regulation of Cav1. 2 calcium channel by microRNA miR-103: role in pain. The EMBO journal 2011; 30(18): 3830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kim DM, Nimigean CM. Voltage-gated potassium channels: a structural examination of selectivity and gating. Cold Spring Harb Perspect Biol 2016; 8(5): a029231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sakai A, Saitow F, Maruyama M, Miyake N, Miyake K, Shimada T, Okada T, Suzuki H. MicroRNA cluster miR-17-92 regulates multiple functionally related voltage-gated potassium channels in chronic neuropathic pain. Nat Commun 2017; 8(1): 16079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhang J, Rong L, Shao J, Zhang Y, Liu Y, Zhao S, Li L, Yu W, Zhang M, Ren X, Zhao Q, Zhu C, Luo H, Zang W, Cao J. Epigenetic restoration of voltage-gated potassium channel Kv1. 2 alleviates nerve injury‐induced neuropathic pain. J Neurochem 2021; 156(3): 367. [DOI] [PubMed] [Google Scholar]
- 90.Lindia JA, Köhler MG, Martin WJ, Abbadie C. Relationship between sodium channel NaV1. 3 expression and neuropathic pain behavior in rats. Pain 2005; 117(1-2): 145. [DOI] [PubMed] [Google Scholar]
- 91.Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The role of voltage-gated sodium channels in pain signaling. Physiol Rev 2019; 99(2): 1079. [DOI] [PubMed] [Google Scholar]
- 92.Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics 2010; 73(10): 1907. [DOI] [PubMed] [Google Scholar]
- 93.Cata JP, Uhelski ML, Gorur A, Dougherty PM. Nociception and pain: new roles for exosomes. Neuroscientist 2022; 28(4): 349. [DOI] [PubMed] [Google Scholar]
- 94.Ding S-Q, Chen J, Wang S-N, Duan F-X, Chen Y-Q, Shi Y-J, Hu JG, Lü HZ. Identification of serum exosomal microRNAs in acute spinal cord injured rats. Exp Biol Med 2019; 244(14): 1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Jiang B-C, Liu T, Gao Y-J. Chemokines in chronic pain: cellular and molecular mechanisms and therapeutic potential. Pharmacol Ther 2020; 212: 107581. [DOI] [PubMed] [Google Scholar]
- 96.Groot M, Lee H. Sorting mechanisms for MicroRNAs into extracellular vesicles and their associated diseases. Cells 2020; 9(4): 1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Torralba D, Baixauli F, Villarroya-Beltri C, Fernández-Delgado I, Latorre-Pellicer A, Acín-Pérez R, Martín-Cófreces NB, Jaso-Tamame ÁL, Iborra S, Jorge I, González-Aseguinolaza G, Garaude J, Vicente-Manzanares M, Enríquez JA, Mittelbrunn M, Sánchez-Madrid F. Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat Commun 2018; 9(1): 2658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kalani A, Tyagi A, Tyagi N. Exosomes: mediators of neurodegeneration, neuroprotection and therapeutics. Mol Neurobiol 2014; 49: 590–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Console L, Scalise M, Indiveri C. Exosomes in inflammation and role as biomarkers. Clin Chim Acta 2019; 488: 165. [DOI] [PubMed] [Google Scholar]
- 100.Ni Z, Kuang L, Chen H, Xie Y, Zhang B, Ouyang J, Wu J, Zhou S, Chen L, Su N, Tan Q, Luo X, Chen B, Chen S, Yin L, Huang H, Du X, Chen L. The exosome-like vesicles from osteoarthritic chondrocyte enhanced mature IL-1β production of macrophages and aggravated synovitis in osteoarthritis. Cell Death Dis 2019; 10(7): 522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ding X, Jing N, Shen A, Guo F, Song Y, Pan M, Ma X, Zhao L, Zhang H, Wu L, Qin G, Zhao Y. MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. J Endocrinol Invest 2021; 44: 1175. [DOI] [PubMed] [Google Scholar]
- 102.Zhang Y, Liu J, Wang X, Zhang J, Xie C. Extracellular vesicle-encapsulated microRNA-23a from dorsal root ganglia neurons binds to A20 and promotes inflammatory macrophage polarization following peripheral nerve injury. Aging (Albany NY) 2021; 13(5): 6752–6764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ren J, Liu N, Sun N, Zhang K, Yu L. Mesenchymal stem cells and their exosomes: promising therapeutics for chronic pain. Curr Stem Cell Res Ther 2019; 14(8): 644. [DOI] [PubMed] [Google Scholar]
- 104.Li C, Li X, Zhao B, Wang C. Exosomes derived from miR-544-modified mesenchymal stem cells promote recovery after spinal cord injury. Arch Physiol Biochem 2020; 126(4): 369. [DOI] [PubMed] [Google Scholar]
- 105.Zhao L, Jiang X, Shi J, Gao S, Zhu Y, Gu T, Shi E. Exosomes derived from bone marrow mesenchymal stem cells overexpressing microRNA-25 protect spinal cords against transient ischemia. J Thorac Cardiovasc Surg 2019; 157(2): 508. [DOI] [PubMed] [Google Scholar]
- 106.Lv B, Zhang X, Yuan J, Chen Y, Ding H, Cao X, Huang A. Biomaterial-supported MSC transplantation enhances cell–cell communication for spinal cord injury. Stem Cell Res Ther 2021; 12: 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Amaechi O, Huffman MM, Featherstone K. Pharmacologic therapy for acute pain. Am Fam Physician 2021; 104(1): 63–72. [PubMed] [Google Scholar]
- 108.Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther 2018; 9: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Liu Y, Wan X, Yuan Y, Huang J, Jiang Y, Zhao K, Wang Y, Liu Y, Wang Q, Jin H. Opposite effects of miR-155 in the initial and later stages of lipopolysaccharide (LPS)-induced inflammatory response. J Zhejiang Univ - Sci B 2021; 22(7): 590–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Arabpour M, Saghazadeh A, Rezaei N. Anti-inflammatory and M2 macrophage polarization-promoting effect of mesenchymal stem cell-derived exosomes. Int Immunopharmacol 2021; 97: 107823. [DOI] [PubMed] [Google Scholar]
- 111.Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008; 8(9): 726. [DOI] [PubMed] [Google Scholar]
- 112.Norte-Muñoz M, Lucas-Ruiz F, Gallego-Ortega A, García-Bernal D, Valiente-Soriano FJ, de la Villa P, Vidal-Sanz M, Agudo-Barriuso M. Neuroprotection and axonal regeneration induced by bone marrow mesenchymal stromal cells depend on the type of transplant. Front Cell Dev Biol 2021; 9: 772223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Xin H, Li Y, Liu Z, Wang X, Shang X, Cui Y, Zhang ZG, Chopp M. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem cells 2013; 31(12): 2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ren Z-W, Zhou J-G, Xiong Z-K, Zhu F-Z, Guo X-D. Effect of exosomes derived from MiR-133b-modified ADSCs on the recovery of neurological function after SCI. Eur Rev Med Pharmacol Sci 2019; 23(1): 52–60. [DOI] [PubMed] [Google Scholar]
- 115.Xin H, Katakowski M, Wang F, Qian J-Y, Liu XS, Ali MM, Buller B, Zhang ZG, Chopp M. MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 2017; 48(3): 747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Cong M, Shen M, Wu X, Li Y, Wang L, He Q, Shi H, Ding F. Improvement of sensory neuron growth and survival via negatively regulating PTEN by miR-21-5p-contained small extracellular vesicles from skin precursor-derived Schwann cells. Stem Cell Res Ther 2021; 12: 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Xia B, Gao J, Li S, Huang L, Zhu L, Ma T, Zhao L, Yang Y, Luo K, Shi X, Mei L, Zhang H, Zheng Y, Lu L, Luo Z, Huang J. Mechanical stimulation of Schwann cells promote peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b-3p. Theranostics 2020; 10(20): 8974–8995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Wang W, Li R. MiR-216a-5p alleviates chronic constriction injury-induced neuropathic pain in rats by targeting KDM3A and inactivating Wnt/β-catenin signaling pathway. Neurosci Res 2021; 170: 255. [DOI] [PubMed] [Google Scholar]
- 119.Ma K, Xu H, Zhang J, Zhao F, Liang H, Sun H, Li P, Zhang S, Wang R, Chen X. Insulin-like growth factor-1 enhances neuroprotective effects of neural stem cell exosomes after spinal cord injury via an miR-219a-2-3p/YY1 mechanism. Aging (Albany NY) 2019; 11(24): 12278–12294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhang X, Liu Z, Shu Q, Yuan S, Xing Z, Song J. LncRNA SNHG6 functions as a ceRNA to regulate neuronal cell apoptosis by modulating miR-181c-5p/BIM signalling in ischaemic stroke. J Cell Mol Med 2019; 23(9): 6120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Le LT, Swingler TE, Crowe N, Vincent TL, Barter MJ, Donell ST, Delany AM, Dalmay T, Young DA, Clark IM. The microRNA-29 family in cartilage homeostasis and osteoarthritis. J Mol Med 2016; 94: 583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang Z, Shen W, Zhu M, Xu M, Qiu M, Zhang D, Chen S. MiR-199-3p suppressed inflammatory response by targeting MECP2 to alleviate RTX-induced PHN in mice. Cell Transplant 2022; 31: 096368972211081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hua T, Yang M, Song H, Kong E, Deng M, Li Y, Li J, Liu Z, Fu H, Wang Y, Yuan H. Huc-MSCs-derived exosomes attenuate inflammatory pain by regulating microglia pyroptosis and autophagy via the miR-146a-5p/TRAF6 axis. J Nanobiotechnology 2022; 20(1): 324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Jin Z, Ren J, Qi S. Human bone mesenchymal stem cells-derived exosomes overexpressing microRNA-26a-5p alleviate osteoarthritis via down-regulation of PTGS2. Int Immunopharmacol 2020; 78: 105946. [DOI] [PubMed] [Google Scholar]
- 125.Hmadcha A, Martin-Montalvo A, Gauthier BR, Soria B, Capilla-Gonzalez V. Therapeutic potential of mesenchymal stem cells for cancer therapy. Front Bioeng Biotechnol 2020; 8: 43. [DOI] [PMC free article] [PubMed] [Google Scholar]



