Next Article in Journal
The ctpF Gene Encoding a Calcium P-Type ATPase of the Plasma Membrane Contributes to Full Virulence of Mycobacterium tuberculosis
Next Article in Special Issue
Alteration of Gene and miRNA Expression in Cervical Intraepithelial Neoplasia and Cervical Cancer
Previous Article in Journal
Kub3 Deficiency Causes Aberrant Late Embryonic Lung Development in Mice by the FGF Signaling Pathway
Previous Article in Special Issue
Histone Modifications and Non-Coding RNAs: Mutual Epigenetic Regulation and Role in Pathogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Expression and Biological Functions of miRNAs in Chronic Pain: A Review on Human Studies

by
Saverio Sabina
1,†,
Alessandra Panico
2,†,
Pierpaolo Mincarone
3,
Carlo Giacomo Leo
1,
Sergio Garbarino
4,
Tiziana Grassi
2,
Francesco Bagordo
5,
Antonella De Donno
2,
Egeria Scoditti
1,* and
Maria Rosaria Tumolo
1,2
1
Institute of Clinical Physiology, National Research Council, Via Monteroni, 73100 Lecce, Italy
2
Department of Biological and Environmental Sciences and Technology, University of Salento, Via Monteroni, 73100 Lecce, Italy
3
Institute for Research on Population and Social Policies, National Research Council, c/o ex Osp. Di Summa, Piazza Di Summa, 72100 Brindisi, Italy
4
Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, 16132 Genoa, Italy
5
Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, Via Edoardo Orabona, 70126 Bari, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(11), 6016; https://doi.org/10.3390/ijms23116016
Submission received: 6 April 2022 / Revised: 24 May 2022 / Accepted: 26 May 2022 / Published: 27 May 2022
(This article belongs to the Collection Regulation by Non-coding RNAs)

Abstract

:
Chronic pain is a major public health problem and an economic burden worldwide. However, its underlying pathological mechanisms remain unclear. MicroRNAs (miRNAs) are a class of small noncoding RNAs that post-transcriptionally regulate gene expression and serve key roles in physiological and pathological processes. This review aims to synthesize the human studies examining miRNA expression in the pathogenesis of chronic primary pain and chronic secondary pain. Additionally, to understand the potential pathophysiological impact of miRNAs in these conditions, an in silico analysis was performed to reveal the target genes and pathways involved in primary and secondary pain and their differential regulation in the different types of chronic pain. The findings, methodological issues and challenges of miRNA research in the pathophysiology of chronic pain are discussed. The available evidence suggests the potential role of miRNA in disease pathogenesis and possibly the pain process, eventually enabling this role to be exploited for pain monitoring and management.

1. Introduction

Chronic pain (CP), an unpleasant sensory and emotional experience that affects many persons worldwide and is a leading chief complaint in ambulatory care settings [1]. It is often the result of damage to and persistent inflammation of peripheral (somatic) or internal (visceral) non-neural tissue (nociceptive pain), or a lesion or disease of the central or peripheral somatosensory nervous system (neuropathic pain) [2]. The International Classification of Diseases (ICD-11) distinguishes two main categories of CP: chronic primary pain (CPP) and chronic secondary pain (CSP). The former persists or recurs for longer than 3 months where the pain can be conceived as a disease and cannot be explained by another chronic condition. CSP appears as a symptom of another disease but is a problem in its own right in many cases, persisting beyond the successful treatment of the initial causing pathology (Table 1) [3]. It is estimated that 20% of European adults have had CP [4]. A similar estimated prevalence has been reported in US adults [5].
CP conditions are identified as a leading cause of global disability [6] regardless of its cause, the body site and the age of the patients. In Europe, its economic burden, in terms of cost per patient per year, has been estimated as between 1800 euros and 10,200 euros, depending on the severity of the CP and the country; its economic weight and long course make it one of the most expensive long-term illnesses for the community [7,8]. CP causes adverse effects on psychological functions (depression, anxiety, coping skills, post-traumatic stress, etc.) as well as on sociocultural aspects (low educational attainment, poor social support, financial barriers or health insurance, substance abuse, etc.), and therefore has negative consequences for individuals, families and society [9].
Current evidence on the management of CP recommends an interdisciplinary approach, including restorative therapies, pharmacotherapies, procedural interventions, behavioral treatments, complementary therapies and integrative therapies [9]. Nonetheless, most patients do not receive specialist pain treatment, and 40% reported inadequate pain management [4]. The absence of optimal therapy is linked to the complexity of CP pathophysiology [10].
Accumulating evidence suggests that the field of epigenetics, i.e., changes in gene expression without modifying genomic sequences, may provide clues for CP management [9]. Among epigenetic mechanisms, microRNAs (miRNAs or miRs) are involved in physiological and pathological processes, and their alteration contributes to a variety of diseases [11,12]. MiRNAs are small noncoding single-stranded RNAs of approximately 22 nucleotides in length that participate in RNA silencing and post-transcriptional regulation of gene expression [13]. They play pivotal roles in many biological processes [14] and have been detected not only inside the cells, but also in extracellular human body fluids [15,16]. Extracellular miRNAs can be packed into apoptotic bodies, microvesicles, exosomes or bond to a protein, so they are highly stable despite the extracellular RNase activities [17]. Due to these properties, miRNAs could be a candidate biomarker in many physiologic and pathologic processes.
There are several studies reporting changes in miRNA expression in musculoskeletal disorders, migraines and other pain conditions. Moreover, a single miRNA can have multiple target genes and regulate many different biological pathways involved in pain processing [18]. Identifying the interactions that occur between miRNAs and their targets is a critical step in defining the regulatory role of miRNAs in the complex networks that regulate biological processes. Manually predicting the interaction of an miRNA with its target is not feasible due to the large and growing number of miRNA species and the huge number of putative targets. In silico analysis using prediction databases is the common method of identifying the potential target genes for miRNAs [19]. Therefore, further understanding of the multiple processes and pathways involved in pain processing [20] and their relationship to miRNA expression could have important clinical implications for CP management.
The purpose of this narrative review is to provide a comprehensive overview of human studies, examining the expression of miRNAs in the pathogenesis of CPP and CSP conditions. In addition, for those dysregulated miRNAs most consistently reported in the reviewed studies as being implicated in CP, bioinformatics analysis was then performed to investigate the dysregulated miRNA target genes and the signaling pathways involved, which may enhance our understanding of the potential pathophysiological impact of miRNAs in CP and of the molecular mechanisms leading to CP.

2. Results

In the following paragraphs, we have described some of the retrieved studies on miRNA expression distinguishing CPP and CSP. No data were found on miRNAs in chronic cancer-related pain, chronic secondary headache or orofacial pain. Table 2 and Table 3 summarize all studies, including information on miRNA extraction and detection methods, study design, sample type and tissue. The nomenclature of miRNAs was reported adopting the most recent official version used in miRbase [21].

2.1. miRNA Expression in Relation to Chronic Primary Pain

2.1.1. Chronic Widespread Pain

Chronic widespread pain (CWP) is diffuse musculoskeletal pain in at least 4 of 5 body regions and in at least 3 or more body quadrants, as well as the axial skeleton [22]. Fibromyalgia (FM) is a form of CWP characterized by a reduced pain threshold and is regularly accompanied by fatigue, sleep disorders and depressive episodes [23]; it is a multifactorial disease whose pathophysiology is not well understood.
Braun et al. showed a downregulation of miR-103a-3p, miR-107 and miR-130a-3p and an upregulation of miR-125a-5p in FM patients compared to a control no-FM group. Moreover, FM patients were grouped according to the phenotypic characteristics of the disease, and only miR-103a-3p was expressed differently among clusters of FM patients. In particular, up-regulated miR-103a-3p and miR-107, which play a regulatory role in inflammation, were associated with adaptive coping in FM patients and the expression of miR-103a-3p correlated with FM-related disability [24].
In another study [25], six circulating miRNAs were significantly and differentially regulated in patients with FM compared to healthy controls. To identify putative gene targets of the deregulated miRNAs, a bioinformatic analysis was conducted, showing that several biological processes related to brain function/development and muscular functions were targeted by the dysregulated miRNAs in FM patients.
Leinders et al. [26] investigated systemic and cutaneous miRNA expression in FM patients. Several miRNAs were differently expressed compared to a control group, and only some of them were validated. Let-7d-5p, miR-103a-3p and miR-146a-5p resulted downregulated in the blood and skin of patients with reduced intraepidermal nerve fiber density (IENFD). Let-7d-5p expression in white blood cells showed positive correlations with IENFD and mean pain scores on the Graded Chronic Pain Scale in FM patients. In skin biopsies, let-7d-5p expression was higher in patients with FM, particularly in the subgroup with reduced IENFD. Furthermore, these patients showed lower expression of skin insulin-like growth factor-1 receptor (IGF-1Rs) than healthy controls. IGFs-1/IGF-1Rs are downstream targets of let-7d-5p (according to bioinformatic analysis) and play a major role in muscle regeneration [27].

2.1.2. Complex Regional Pain Syndrome

Complex regional pain syndrome (CRPS) is characterized by allodynia, hyperalgesia, abnormal motor function and trophic disturbances [22].
A key feature of CRPS is neurogenic inflammation, and in fact in the study of Orlova et al., 14 differentially expressed miRNAs in patients with CRPS correlated with inflammatory and immune related markers, including vascular endothelial growth factor (VEGF), interleukin1 receptor antagonist (IL1Ra) and monocyte chemotactic protein-1 (MCP-1). VEGF and ILR1a, in particular, showed a significant correlation with the patients’ reported pain levels [28]. Douglas et al. showed that the miRNA profile was different between responders and poor responders both before and after ketamine therapy, an N-methyl-D-aspartate receptor agonist currently used for the treatment of CRPS. Among the differentially expressed miRNAs, miR-548d-5p, which was reduced more in poor responders than responders, showed two potential targets, namely UGT1A1 and CYP3A4, both important in drug metabolism. The functional analyses confirmed only UGT1A1, a member of the UDP-glucuronosyltransferase (GT) family. Moreover, the poor responders had a higher percentage of bilirubin than the responders, indicating that they could have increased levels of conjugated inactive glucuronides and therefore a greater removal of ketamine metabolites, potentially minimizing the therapeutic efficacy of ketamine [29]. Another dysregulated miRNA in ketamine-treated CRPS patients was miR-34a-5p, a miRNA with a known role in inflammation. In the study by Shenoda et al., an inverse correlation was observed between this miRNA and the long noncoding RNA X-inactive-specific transcript (XIST) in CRPS patients who responded poorly to therapy. Therefore, a decrease in miR-34a-5p could lead to an upregulation of XIST, which promotes inflammation via the pro-inflammatory transcription factor yin yang 1 [30].
Plasma exchange is an extracorporeal procedure used to purify the blood from molecules that can cause inflammation. In this regard, analysis of miRNAs in exosomes from the serum of responders and poor responders to plasma exchange showed that 17 miRNAs were significantly altered before and after plasma exchange. Among these miRNAs, miR-338-5p was down-regulated in poor responders compared to responders. The miR-338-5p target, i.e., IL-6 (a pro-inflammatory cytokine), was more reduced in responders following plasma exchange than before the therapy, thus directly contributing to the reduction of inflammation in CRPS. These results indicated that the combined approach of miRNA evaluation with cytokine analysis may be a feasible approach that can help predict the therapeutic efficacy of plasma exchange [31].

2.1.3. Chronic Primary Headache or Orofacial Pain

A chronic primary headache or orofacial pain is defined as a headache or orofacial pain that occurs on at least 15 days per month for longer than 3 months and includes several subtypes [22].
Among them, trigeminal neuralgia (TN) is caused by peripheral nerve lesions and adversely affects the life of patients [32]. To date, only one study investigated the potential functions of miRNAs in human subjects with TN, showing an up-regulation of miR-132-3p, miR-146b-5p, miR-155-5p and miR-384 in patients with TN compared to healthy controls. Bioinformatic analysis revealed the potential involvement of these miRNAs in the onset and development of neuropathic pain. This result was confirmed by functional analysis indicating that miR-155-5p can directly target and downregulate nuclear factor-E2 related factor 2 (Nrf2), a factor that modulates the expression of genes with an implication in inflammation, immune response and antioxidant actions, all mechanisms involved in the development of TN [33].
Migraines, a disabling neurovascular disorder common in adults and children, is associated with activation and sensitization of the trigeminal pain circuit, vascular endothelial cells, inflammation and most likely dysregulation of glial cell activity [34].
Cheng et al. [35] conducted a study to evaluate the level of specific endothelial miRNAs in migraine patients. The miRNAs analyzed were chosen on the basis of their implication in the modulation of inflammation and oxidative stress in endothelial cells [36,37]. MiR-126-3p, miR-155-5p and let-7g-5p were found to be overexpressed in the migraine patients, while a similar but non-significant trend was observed for miR-21-5p. A correlation of miR-155-5p and miR-126-3p with syncope attack suggested that migraines could be a risk factor for syncope. In addition, the level of an endothelial dysfunction marker (intercellular adhesion molecule 1) showed a positive correlation with miR-155-5p, miR-21-5p and let-7g-5p in migraine patients. Altogether, these findings point out that migraine and vascular events may be associated with endothelial dysfunction [35].
In the study by Gallelli et al., overexpression of miR-34a-5p and miR-375 was found in young subjects with migraines and without aura who were not undergoing drug treatment, compared to the treated group and healthy subjects. The in-silico analysis showed that the target genes, i.e., HCN3, NAV3, GPR158 for miR-34a-5p and MTPN for miR-375, are involved in trigeminal pain circuit of migraine [38].
Another problem in the evolution of migraines is the chronification that leads to increased disability. In this case, associated medication overuse could lead to reduced benefits. To study biomarkers involved in pathophysiology and the chronification of migraines, Greco et al. evaluated the expression of miR-34a-5p and miR-382-5p as well as that of calcitonin gene-related peptide (CGRP). The latter, due to its functions in cell degranulation, neurogenic inflammation and vasodilation, is involved in migraine headaches. The miRNAs analyzed were up-regulated in chronic migraine subjects with medication overuse compared to the episodic migraine group, and were positively correlated with CGRP levels. Moreover, the miRNA profile studied after detoxification from overused drugs showed reduced expression, in parallel with similar reduction also observed in CGRP levels. Hence, the medication overuse can modify the biology of migraine, including miRNA expression, and CGRP can play a role in migraine chronification [39].

2.1.4. Chronic Primary Visceral Pain

Chronic primary visceral pain is a CPP localized in the head or neck, thoracic, abdominal or pelvic region, and is associated with related specific internal organs [22].
Irritable Bowel Syndrome (IBS) is a common chronic functional gastrointestinal disorder characterized by chronic abdominal pain and altered bowel habits, including diarrhea (IBS-D), constipation (IBS-C) or a mixture of both. IBS patients may suffer from visceral hypersensitivity and elevated visceral nociception [40], and several neurotransmitters are implicated in these processes, such as transient receptor potential vanilloid type 1 (TRPV1), serotonin reuptake transporter (SERT), etc. [41]. According to these premises, Zhou et al. found reduced expression of miR-199a-5p and miR-199b-5p in patients with IBS-D, which was related to the overexpression of TRPV1, a cation channel that, when activated, produces a burning sensation and/or pain [42]. Thus, the expression of these two miRNAs may contribute to chronic visceral pain and nociception through the up-regulation of TRPVI [41].
Another feature of IBS is the impaired intestinal mucosal barrier function, in which tight junctions and transmembrane proteins, including the cytosolic tight junction protein ZO-1 and claudin-1 (CLDN1), play an important role. In this regard, Zhu et al. investigated whether miR-29a-3p (a miRNA with potential implication in IBS) is associated with ZO-1 and CLDN1 and thus regulates intestinal mucosal barrier function. The level of miR-29a-3p was higher in patients with IBS-D than in healthy subjects, while the expression of ZO-1 and CLDN1 was lower, suggesting their potential role in the pathogenesis of IBS [43]. Another study on the association of intestinal barriers in IBS with miRNA expression was conducted by Mahurkar-Joshi et al., which found reduced expression of miR-219a-5p and miR-338-3p in IBS and IBS-C compared to healthy controls. The functional analysis showed that miR-219a-5p and miR-338-3p potentially contributed to altering barrier function and visceral hypersensitivity through dysregulated expression of proteasome/barrier function genes and of Mitogen Activated Protein Kinase (MAPK) signaling pathway genes, implicated in inflammation pain and hypersensitivity [44].

2.1.5. Chronic Primary Musculoskeletal Pain

Chronic primary musculoskeletal pain (CPMP) is chronic pain experienced in muscles, bones, joints or tendons that cannot be directly attributed to a known disease or a damage process [22]. Among CPMP, multidisciplinary treatment programs are recommended as a first-line treatment for low back pain (LBP), given its multifactorial causes [45]. In this regard, the expression profiles of some miRNAs were evaluated in patients with chronic LBP and compared with those of healthy volunteers before the start of a multidisciplinary treatment program. Significant upregulation of miR-124-3p, miR-150-5p and miR-155-5p was found in patients with chronic LBP comparted to controls. Furthermore, the expression of these miRNAs was analyzed before and after treatment in responders and non-responders, and only the expression of miR-124-3p significantly increased in patients who responded to treatment [46]. MiR-124-3p regulates neurogenesis, neuronal differentiation and stress response [47], and is also considered one of the most important miRNAs for psychological disorders [48]. Therefore, according to the authors, the improvement in miR-124-3p expression may reflect the positive effect of multidisciplinary treatment on both pain and stress [46].
Table 2. MiRNA dysregulation related to chronic primary pain.
Table 2. MiRNA dysregulation related to chronic primary pain.
Type of PainmiRNAsmiRNA Expression Extraction MethodDetection MethodStudy DesignSample SizeTissueReference
Chronic widespread pain
Fibromyalgia103a-3p, 107, 130a-3p
125a-5p

miRNeasy Mini kit (Qiagen, Germany)qRT-PCRCross-sectional31 FM
16 HC
Blood[24]
21-5p, 23a-3p, 23b-3p 29a-3p, 99b-5p, 125b-5p, 145-5p, 195-5p, 223-3pmiRNeasy Mini kit (Qiagen, Germany)qRT-PCRCross-sectional10 FM
8 HC
Cerebrospinal fluid[49]
let-7a-5p, 30b-5p, 103a-3p, 107, 151a-5p, 142-3p, 374b-5p
320a-3p

miRNeasy Mini kit (Qiagen, Germany)qRT-PCRCross-sectional20 FM
20 HC
Serum [50]
143-3p, 145-5p, 223-3p, 338-3p, 451a mirVana miRNA isolation kitqRT-PCRCross-sectional11 FM
10 HC
PBMCs[51]
1-3p, 23a-3p, 133a, 139-5p, 320b, 346miRCURY RNA Isolation Kit-BiofluidsqRT-PCRCross-sectional14 FM
14 HC
Serum[25]
let-7d-5p, 103a-3p, 146a-5pRNeasy Micro kit (Qiagen, Germany)qRT-PCRCross-sectional30 FM (12 reduced IENFD)
34 HC
Skin[26]
CRPSlet-7a-5p, let-7b-5p, let-7c, 25-5p, 126-3p, 181a-2-3p,320a-3p, 320b, 532-3p, 625-5p, 629-5p, 664a-3p,
939-5p, 1285-3p
PAXgene blood miRNA kit (Qiagen, Valencia, CA, USA)TLDACross-sectional41 FM
20 HC
Whole blood[28]
33 miRNAs (before treatment)
43 miRNAs (after)
DysregulatedPAXgene blood miRNA kit (Qiagen, Hilden, Germany)TLDACross-sectional7 Responders Ketamine, 6 PR KetamineWhole blood[29]
34a-5pPAXgene blood RNA tubes (BD Biosciences, Franklin Lakes, NJ, USA)qRT-PCRCross-sectional7 Responders Ketamine, 6 PR KetamineWhole blood[30]
Let-7a-5p, 19b-3p, 19b-1-5p, 29b-3p, 30a-5p, 34b-5p, 126-5p, 191-5p, 195-5p, 338-5p, 484, 518b, 542-3p, 551b-5p, 618, 1183, 1274bDysregulatedmiRvana miRNA isolation kit (Life technologies)TLDARetrospective case series3 RE-PE, 3 NR-PESerum[31]
Chronic primary headache or orofacial pain
Trigeminal neuralgia132-3p, 146b-5p, 155-5p, 384TRIzol reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional28 TN
31 HC
Serum[33]
Migrainelet-7g-5p, 126-3p, 155-5p miRNeasy Serum/ Plasma Kit (Qiagen, Germany)qRT-PCRCross-sectional30 MP
30 HC
Plasma[35]
27b-3p
let-7b-5p, 22-3p, 181a-5p

mirCURY LNATM Universal cDNA syn- thesis kit (Exiqon)qRT-PCRCross-sectional15 MP
13 HC
PBMC[52]
34a-5p, 375miRNeasy Serum/Plasma Kit (Qiagen, Venlo, The Netherlands)qRT-PCRProspective parallel-group12 untreated- MPWA
12 HC
Saliva[38]
12 untreated- MPWA
12 treated-MPWA
Saliva and serum
30a-5p↑ in migraineTRIzol reagent (TaKaRa, Otsu, Shiga, Japan)qRT-PCRCross-sectionalNot reportedSerum[53]
34a-5p, 382-5p
Direct-zol RNA Mini prep plus (Zymo Research from Aurogene, Rome, Italy)qRT-PCRCross-sectional observational controlled 28 CM-MO
27 EM
PBMC[39]
29c-5p, 34a-5p, 382-5pRNeasy® Plus 96 Kit (Qiagen, Hilden, GermanyqRT-PCRCohortCohort1: 8 MPA
8 MPPF
Serum[54]
29c-5p, 382-5p Cohort1: 8 MPPF
8 HC
382-5p miRNeasy® Serum/Plasma Kit (Qiagen, Hilden, Germany) Cohort2: 12 MPPF 12 HC
Chronic primary visceral pain
Irritable bowel syndrome199a-5p, 199b-5pTrizol (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional45 IBS-D
40 HC
Colon tissue[41]
24-3pmiREasy Mini Kit Qiagen, GermanyqRT-PCRCross-sectional10 IBS-D
10 HC
Intestinal mucosa epithelial cells[55]
29a-3pmiRcute miRNA isolation kit (Tiangen Biotech Co., Ltd.)qRT-PCRCross-sectional21 IBS-D
16 HC
Sigmoid colon mucosa[43]
106b-5p, 532-5p
363-3p, 338-3p (IBS vs HC)
106b-5p
100-5p, 338-3p (IBS-C vs HC)
219-5p (IBS-D vs HC)




Trizol (Invitrogen, Carlsbad, CA, USA)nanoString nCounter Assay Cross-sectional29 IBS (15 IBS-C, 14 IBS-D)
15 HC
Colon tissue[44]
219-5p, 338-3p (IBS and IBS-C vs HC) qRT-PCR
Chronic primary musculoskeletal pain124-3p, 150-5p, 155-5pmirVana miRNA Isolation Kit (Ambion, Austin, TX, USA)qRT-PCRProspective cohort34 LBP
20 HC
CD4+ cells[46]
Legend: CM-MO: chronic migraine-overuse of medications; EM: episodic migraine; HC: healthy controls; IBS-C: Irritable bowel syndrome-constipation; IBS-D: Irritable bowel syndrome- diarrhea; IC: interstitial cystitis; IENFD: intraepidermal nerve fiber density; LBP: low back pain; MP: Migraine patients; MPWA: Migraine patients without aura; MPA: migraine patients attack; MPPF: migraine patients pain-free; NR-PE: non-responders plasma exchange; PR: poor responder; qRT-PCR: Quantitative real-time PCR; RE-PE: responder plasma exchange; TN: trigeminal neuralgia. ↓/↑, difference in miRNAs expression, respectively up- and down-expressed.

2.2. miRNA Expression in Relation to Chronic Secondary Pain

2.2.1. Chronic Post-Surgical Pain or Post-Traumatic Pain

One of the most common symptoms after major surgery is pain that, if undertreated, can lead to the development of persistent postoperative pain, increasing the risk of opioid addiction and related adverse effects [56]. The nociceptive and neuropathic components, often concomitant, are involved in the pathophysiology of postoperative pain. Human evidence specifically related to CSP to surgery is very scarce.
The exploratory study by Giordano et al. evaluated the potential use of circulating miRNAs as predictive biomarkers for chronic postoperative pain. The preoperative expression of circulating miRNAs, as validated or predicted to be associated with pain, inflammation and matrix degeneration, was measured in patients with severe knee osteoarthritis (OA) scheduled for total knee replacement and classified into two groups, the high-pain relief group and the low-pain relief group, based on the percentage of postoperative pain relief one year after surgery (more than 30% pain relief and less than 30% pain relief, respectively). Three miRNAs, miR-146a-5p, miR-145-5p and miR-130b-3p, were significantly dysregulated in the low-pain relief group compared to the high-pain relief group, and miR-146a-5p tended to be an independent predictor of postoperative pain relief [57]. Therefore, postoperative pain appeared to be associated with a specific preoperative serum miRNA signature.
The involvement of the inflammatory/oxidative stress modulatory miR-21-5p in post-surgical pain has been proposed in the context of surgical removal of the impacted mandibular third molar [58], where increased bone expression of miR-21-5p has been associated with a delayed postoperative pain onset, potentially reflecting a reduction in surgical (and inflammatory) stress. However, this study addressed short-term surgical outcomes including pain, and the role for miR-21-5p in chronic post-surgical pain needs to be determined.
Prospective studies have indicated that miRNA profiles can serve as molecular and blood-based predictors of chronic post-traumatic musculoskeletal pain. Indeed, 32 miRNAs from blood collected following a traumatic/stressful event such as a motor vehicle collision were significantly different among individuals who did and did not develop severe persistent musculoskeletal pain in the axial region (neck, shoulders, and/or back) 6 weeks after the trauma [59]. In particular, two miRNAs, miR-135a-5p (regulating serotonin receptor and transporter) and miR-3613-3p (regulating pain-associated genes), strongly predicted the subsequent development of CP. Most of these differentially expressed miRNAs are associated with stress response and/or pain processing, thus supporting their role in the pathogenesis of CP. Interestingly, the differentially expressed miRNAs were enriched for gene location on the X chromosome and appeared to contribute to persistent severe axial pain in women but not in men [59]. The same authors demonstrated that reduced levels of circulating miR-320a-3p in the peritraumatic period predicted persistent diffuse and axial musculoskeletal pain 6 months after a motor vehicle collision in subjects suffering from significant peritraumatic distress, possibly targeting stress and pain-associated transcripts, as demonstrated by bioinformatic analysis and in vitro luciferase reporter assays [60]. Furthermore, peritraumatic circulating levels of miR-19b-3p, which target pain and circadian rhythm related genes, also predicted widespread posttraumatic pain and posttraumatic stress symptoms at follow-up in a sex-dependent manner [61]. Indeed, an inverse relationship between miR-19b-3p expression levels and the probability of developing posttraumatic widespread pain and stress symptoms was shown in women, and a positive relationship between miR-19b-3p expression and the probability of developing these outcomes was shown in men.
Another prospective study was conducted on the whole blood of individuals who reported neck pain related to motor vehicle collisions. Biologic pathways targeted by pain-associated miRNAs include extracellular matrix receptor interaction, TGF-β signaling, amphetamine addiction and axon guidance, all contributing to stress, pain pathogenesis and persistent posttraumatic disability related to neck pain. Furthermore, let-7i-5p expression, previously associated with adipose tissue function, appeared to mediate the association between neck muscles fat infiltration and neck-pain related disability [62].

2.2.2. Chronic Neuropathic Pain

Chronic neuropathic pain (CNP), as previously mentioned, is typically perceived within the innervation territory, which is somatotopically represented within the structure of the damaged nervous system [3].
Several studies have examined the signature of miRNAs in CNP. Liu et al. [63] reported the downregulation of miR-101-3p in both plasma and sural nerves of subjects with neuropathic pain of various origins. Suppression of miR-101-3p has been correlated with pain pathophysiology through inhibition of miR-101-3p-mediated nuclear factor kappa B (NF-κB) activation, a key contributor to the neuroinflammation involved in the pathogenesis of CNP [64]. In accordance with the role of inflammation in the pathogenesis of CP in peripheral neuropathies, inflammation-associated miRNAs were found to be specifically deregulated in peripheral neuropathies of different etiology in comparison with healthy controls, with miR-21-5p (a key regulator of immunological processes) up-regulated in white blood cells and sural nerve especially in painful neuropathies, miR-146a-5p (with anti-inflammatory action) up-regulated in white blood cells and down-regulated in distal skin especially in painful neuropathies and miR-155-5p (otherwise known as pro-inflammatory) down-regulated in white blood cells and distal skin especially in painful inflammatory neuropathies and up-regulated in the sural nerve of painful neuropathies. Strikingly, the aberrant nerve miRNA expression correlated with pain in peripheral neuropathies [65]. Taken together, these results suggest a complex reciprocal neuro-immune interaction at both the systemic and local levels, in which miRNAs may play an active modulating role.
Patients in the acute phase of Herpes Zoster infection compared to controls showed 6 significantly up-regulated miRNAs (miR-190b, -571, -1276, -1303, -661, -943) in serum samples, which were related to the regulation of immunity and inflammation as well as nervous system development. About one-tenth of patients with acute Herpes Zoster develop post-herpetic neuralgia, which is attributed to sensory nerve damage and may persist for at least 3 months [66].
Intervertebral disc degeneration (IDD) is a common degenerative disease whose main manifestations are lower back pain and lumbar disc herniation (LDH), which weigh heavily on the global health system. The pathogenesis of IDD involves a pro-inflammatory reaction with recruitment and activation of immune cells, release of inflammatory factors including IL-6 and TNF-α, extracellular matrix degradation, cell apoptosis, altered autophagy, vascular proliferation and nerve cell degeneration, which leads to nerve root sensitization and CNP [67].
In the study by Cui et al., the signature of circulating miRNAs of patients with IDD and manifest LDH was analyzed. A total of 73 dysregulated miRNAs resulted compared to a control group; among them, upregulated miR-766-3p and miR-6749-3p and downregulated miR-4632-5p can target multiple genes related to IDD and LDH pathogenesis. In particular, bioinformatic analysis identified many pathways, such as endocytosis, apoptosis, axon guidance, vascular endothelial growth factor (VEGF), cell adhesion molecular, focal adhesion, extracellular matrix (ECM)-receptor interaction, phosphoinositide 3-kinase (PI3K)/Akt, mTOR and NF-κB pathways, showing the involvement of differentially expressed miRNAs in the modulation of inflammation, proliferation, differentiation and oxidative stress [68].
The pathogenesis of IDD was studied by Dong et al. [69], who collected nucleus pulposus (NP) and annulus fibrosus cells (which are intervertebral disc components) from patients with IDD; patients with idiopathic scoliosis were used as a control. MiR-640 was significantly upregulated only in NP cells from patients with IDD. In vitro experiments showed that inflammatory cytokines (TNF-α and IL-1β) were also elevated in NP cells, stimulating miR-640 expression via the NF-κB signaling pathway. This may generate a positive feedback loop to aggravate inflammation in IDD, as a high level of miR-640 enhanced TNF-α and IL-1β production. MiR-640 also repressed the Wnt signaling pathway, which has important functions during senescence and apoptosis, suggesting that this miRNA is implicated in the degeneration of NP cells and could be used as a potential therapeutic target for the biotherapy of low back pain.
Given the neuropathic and neuroinflammatory nature of disc herniation-related leg pain, Hasvik et al. investigated the expression of miR-17-5p belonging to the miR-17-92 cluster and influencing the development of neuropathic pain conditions. A total of 97 patients were followed up for 1 year after disc herniation, and elevated serum miR-17-5p expression was shown to be associated with increased leg pain intensity. After an in vivo experiment using a rat model, it has been suggested that the high expression of miR-17-5p in the CNP facilitates the local release of TNF from circulating immune cells via a pro-inflammatory mechanism, which increases pain intensity at the legs after disc herniation [70].

2.2.3. Chronic Secondary Visceral Pain

Chronic secondary visceral pain is a persistent or recurrent pain that originates from internal organs of the head/neck region and the thoracic, abdominal and pelvic cavities. It includes bladder pain syndrome (BPS), chronic prostatitis, endometriosis, etc. [3].
BPS is a clinical syndrome of pelvic pain and urinary frequency and urgency without infection. A possible cause of BPS can be epithelium dysfunction resulting in leakage of the bladder urothelium. Several molecules are involved in the regulation of epithelial permeability and bladder contractility and inflammation, and their downregulation may indicate impaired tight junction structure and increased permeability of the bladder urothelium [71]. Among these down-regulated molecules, Sanchez Freire et al. [72], in their study, showed a significantly lower expression of neurokinin 1 (NK1) tachykinin receptor in BPS patients than in controls. Moreover, miR-499b-5p, miR-500a-5p, miR-328-3p and miR-320a-3p were upregulated in biopsies of BPS patients and negatively correlated with NK1R mRNA and/or protein levels, indicating that BPS can lead to an attenuation of NK1 tachykinin receptor synthesis via activation of these miRNAs [72]. In the study by Arai et al. [73], small RNA sequencing performed on interstitial cystitis tissues, normal bladder tissue and bladder cancer revealed the down-regulation of miR-320 family in interstitial cystitis tissues. The in silico analysis carried-out on this miR-family showed 85 putative targets, but the authors focused on three transcription factors, namely E2F transcription factor 1, E2F transcription factor 2 and tubby bipartite transcription factor, due to their potential to significantly influence downstream RNA networks. The immunohistochemistry assay on these proteins displayed greater expression in interstitial cystitis cells than in normal bladder and bladder cancer tissues. Considering the functions of these three proteins, their overexpression could lead to the inflammation observed in bladder pain syndrome [73].
Prostatitis is another visceral pain syndrome made up of 4 main categories: non-bacterial acute prostatitis, bacterial chronic prostatitis, chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) and asymptomatic inflammatory prostatitis [74]. The study by Chen et al. demonstrated the presence of deregulated miRNAs in the prostatic secretion of CP/CPPS patients, and miR-21-5p, miR-103a-3p and miR-141-3p were further validated, demonstrating that miR-21-5p possessed a high classify-accuracy for CP/CPPS patients with a significant pain score [75].
Regarding endometriosis, a gynecological disease characterized by the presence of endometrial-like tissue outside the uterus that induces inflammatory reactions, in the study by Liang et al. [76], miR-200c-3p was found to be downregulated in ectopic endometrial tissues compared to normal endometrial tissue. Furthermore, the expression of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was upregulated in ectopic endometrial tissues compared to normal endometrial tissues, and the level of MALAT1 was negatively correlated with the level of miR-200c-3p. Thus, miR-200c-3p may suppress the proliferation and migration of endometrial stromal cells by downregulating MALAT1, which in turn functions as a competing endogenous RNA to upregulate the expression of Zinc Finger E-Box Binding Homeobox 1 and 2 by sequestering miR-200c-3p. The MALAT1/miR-200c sponge could act as a novel target for the diagnosis and treatment of endometriosis [76].
Razi et al. compared the plasma level of miR-185-5p in women with endometriosis and a control group. This study showed that there was significantly lower expression of miR-185-5p in the plasma of the case group compared to controls, but no significant difference in the expression of VEGF and PDGF (platelet-derived growth factor) mRNAs, two proangiogenic factors involved in endometriosis and predicted targets of miR-185-5p. Therefore, circulating VEGF and PDGF levels appear to be unrelated to endometriosis, while miR-185-5p could potentially serve as a candidate biomarker for endometriosis [77].

2.2.4. Chronic Secondary Musculoskeletal Pain

Chronic secondary musculoskeletal pain is a chronic musculoskeletal pain that arises from an underlying disease [78]. The study by Dayer et al. [79] showed that some miRNAs were expressed differently depending on the origin of the pain. 10 miRNAs were validated in patients diagnosed with CP of either neuropathic or nociceptive origin. Among these, 7 miRNAs showed significant differential expression between the nociceptive and neuropathic groups. Moreover, miR-320a and miR-98-5p were able to discriminate between the two groups of CP, thus suggesting the potential of these miRNAs as diagnostic tools. Regarding the biological significance of these miRNA changes in CP, some speculations have been made by the authors, including the role in inflammation for miR-98 and miR-205, and specifically in neuropathic pain for let-7d, miR-29c and miR-320, as previously demonstrated [79].
Osteoarthritis (OA), the most common form of arthritis, is a chronic painful disease of the synovial joints associated with cartilage destruction and inflammation leading to motor disability [80]. The mechanism’s underlying pain in OA are multifactorial, including nociceptive, inflammatory and neuropathic components, and are patient- and time-specific, thus complicating the treatment strategy [81]. Angiogenesis, the formation of new blood vessels from pre-existing ones, can promote synovial inflammation and pain, and, in fact, angiogenesis inhibitors have been shown to reduce pain in OA models [82]. According to this premise, Xie et al. [83] found that the angiogenesis-related miR-210, known to regulate the expression of the pro-angiogenic factor VEGF, was upregulated in synovial fluid samples from patients with both early- and late-stage OA compared to healthy subjects, in correlation with the increased protein expression of VEGF. Although further validation is required, the early change in miR-210 expression in OA suggests its potential as an early diagnostic biomarker for OA.
In another study, the expression of 18 miRNAs potentially associated with OA pathology in serum and cartilage samples obtained from patients with OA was examined [84]. Among the tested miRNAs, miR-146a-5p expression was significantly upregulated in both tested OA patients’ samples compared to healthy controls, with a positive correlation between levels in both sample types. This finding suggests that miR-146a-5p serum levels could reflect molecular processes in cartilage, thus potentially representing a biomarker for OA [84]. These results are in line with previous data documenting an increase in the expression level of miR-146a-5p in the cartilage of patients with OA, its induction under inflammation and the reported role for this miRNA in OA pathogenesis and pain through the regulation of genes involved in synovial inflammation, neoangiogenesis and osteoclastogenesis.
MiR-34a-5p, another miRNA associated with cellular processes linked to the pathogenesis of OA, such as inflammation, cell growth and apoptosis, was found to be up-regulated in cartilage samples as well as inflamed primary chondrocytes from patients with OA compared to controls [85]. Its induction has been associated with inflammation-related down-regulation of the long non-coding RNA SNHG7 (small nucleolar RNA host gene 7), which targets miR-34a-5p. As a consequence of the upregulation of miR-34a-5p, the expression of its target synoviolin 1 (SYVN1), which is involved in endoplasmic reticulum stress, chronic inflammation and vascular overgrowth, was reduced [85]. Although the precise role of miR-34a-5p in pain development and/or maintenance is unknown, it shows several pain-relevant targets, including SCN2B, KCNK3, CACNA1E and soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptors (SNAREs) such as VAMP-2, syntaxins and SNAP-25, some of which have been shown, at least in animal models, to be potentially involved in neuropathic [86] and inflammatory pain [87].
Recently, a molecular mechanism in OA has been proposed from clinical and in vitro evidence in which extracellular matrix degradation and chondrocytes apoptosis have been associated with upregulation of the transcription factor lymphoid enhancer-binding factor 1 and the subsequent induction of the circular RNA circRNF121, a non-coding regulatory RNA that targets miR-665, thereby releasing its inhibition on the MYD88/NF-κB pro-inflammatory pathway. Concordantly, reduced levels of miR-665 were found in the cartilage tissues of patients with OA compared to normal tissues. Although the role of miR-665 in OA-related inflammation and degenerative processes has been postulated in this study [88], its function in OA-associated pain processes remains unclear.
A recent prospective study looked for potential prognostic miRNAs based on the change in miRNA expressions after elevated tibial osteotomy in the medial OA compartment of the knee. Two miRNAs, miR-23a-3p and miR-30c-5p, were significantly altered after high tibial osteotomy compared to the preoperative period. Changes in clinical symptoms and radiological measurements were then used to identify specific miRNAs related to better outcomes. Only miR-30c-5p showed significant correlation with the postoperative pain relief [89]. Although the clinical implications and related mechanisms of these changes in miRNAs expression remain to be conclusively demonstrated, the results indicate the potential of these miRNAs as biomarkers for clinical improvement after a high tibial osteotomy.
Table 3. MiRNA dysregulation related to chronic secondary pain.
Table 3. MiRNA dysregulation related to chronic secondary pain.
Type of PainmiRNAmiRNA ExpressionExtraction MethodDetection MethodStudy DesignSample SizeTissueReference
Post-surgical pain130b-3p, 145-5p, 146a-5p↓ (low-pain relief group vs high-pain relief group)miRNeasy Serum/Plasma Advanced Kit (QIAGEN, Hilden, Germany) qRT-PCRProspective (1 year)136 patients with a knee OA scheduled for total knee replacement, divided at follow-up in 22 low-pain relief group, and 114 high-pain relief groupSerum[57]
Post-traumatic pain135a-5p, 3613-3p, 19b-3p, 502-3p
7-5p, 26b-3p, 185-5p

PAXgene blood miRNA kit (Qiagen, Valencia, CA, USA)qRT-PCRProspective (6 weeks)53 subjects with a motor vehicle collision, who developed (27) or not (26) persistent axial pain at follow-up Whole blood[59]
320a-3p↓ (according to presence/severity of persistent axial and widespread musculoskeletal pain)PAXgene blood miRNA kit (Qiagen)Sequencing and qRT-PCRProspective (6 weeks)69 subjects with a motor vehicle collision, assessed for presence/severity of different axial and widespread musculoskeletal pain at follow-upWhole blood[60]
19b-3p↓ (women)
↑ (men)
PAXgene blood miRNA kit (Qiagen, Germantown, MD, USA)Sequencing and qRT-PCRProspective (6 weeks)179 subjects with a motor vehicle collision, assessed for persistent posttraumatic widespread pain at follow-upWhole blood[61]
14 miRsDeregulatedPAXgene blood microRNA kit (Qiagen, Germantown, MD, USA)Sequencing and qRT-PCRProspective (1 year)43 subjects with a motor vehicle collision, with different neck pain outcomes during follow-upWhole blood[62]
Chronic Neuropathic pain101-3pTrizol Reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional32 CNP
10 HC
Plasma and sural nerve biopsies[63]
132-3pmiRNEASY kit (Qiagen, Hilden, Germany)qRT-PCRCross-sectional30 patients with peripheral neuropathy, 81 patients with painful or painless inflammatory or non-inflammatory neuropathies
30 HC
White blood cells and sural nerve biopsies[90]
21-5p, 146a-5p, 155-5pDeregulatedNot reportedqRT-PCRCross-sectional76 patients with peripheral neuropathies (of which: 24 with inflammatory neuropathy, 31 with non-inflammatory neuropathy, 21with neuropathy of unknown etiology, 39 with a painful neuropathy and 37 with painless neuropathy)
30 HC
White blood cells, sural nerve, and skin punch biopsies[65]
190b, 571, 1276, 1303, 661, 943TRIzol reagent (Invitrogen, San Diego, CA, USA)TLDA and qRT-PCRCross-sectional41 acute Herpes Zoster patients, 35 HCSerum [66]
34c-5p, 107, 127–5p, 486–3p, 892bTrizol Reagent (Invitrogen, Carlsbad, CA, USA)TLDA and qRT-PCRCross-sectional37 patients with acute Herpes Zoster, 29 patients with post-herpetic neuralgiaSerum[91]
223-3pmiRNeasy isolation kit (Qiagen)qRT-PCRProspective (1 year)97 patients with lumbar radicular pain after disc herniation Serum[92]
21-5pTrizol Reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional10 patients with lumbar disc herniation accompanied by nerve root pain, 10 patients with lumbar burst fractures (HC) Nucleus pulposus tissues of intervertebral disc herniation[93]
Intervertebral disc degeneration17 miRNAs
56 miRNAs

TRIzol® reagent (Invitrogen, Carlsbad, CA, USA)SequencingCross-sectional10 IDD
10 HC
Serum[68]
130b-3p, 200c-3p
10a-5p, 25-3p, 34a-5p, 182-5p

miRNeasy mini kit (Qiagen, Valencia, CA, USA)qRT-PCRCross-sectional3 IDD
3 HC
Disc[94]
640TRIzol (Invitrogen, 15596018)qRT-PCRCross-sectional15 IDD
5 HC
NP and AF cells[69]
185-5pmiRNeasy Mini kit (Qiagen GmbH)qRT-PCRCross-sectional10 IDD
10 HC
NP cells[95]
17-5pmiRNeasy serum plasma isolation kit (Qiagen, Hilden, Germany)qRT-PCRCohort97 patients with lumbar radicular leg pain and disc herniationSerum[70]
Chronic secondary visceral pain
Bladder syndrome320-a-3p, 328-3p, 499b-5p, 500a-5p mirVana miRNA isolation kit (Ambion)TLDACross-sectional8 BPS
4 HC
Dome bladder biopsies[72]
320a-3p, 320b, 320c, 320dTRIzol reagent (Invitrogen, Carlsbad, CA, USA)SequencingCross-sectional8 IC cases, 5 normal bladder, 5 bladder casesIC tissues[73]
Prostatitis21-5p, 103a-3p, 141-3pmiRCURY RNA isolation kit (Exiqon, Woburn, MA, USA)qRT-PCRProspective study21 IIIA CP/CPPS patientsProstatic secretion[75]
Endometriosis185-5pQIAamp miRNA Plasma/Serum Mini kit (QIAGEN, Germany)qRT-PCRCase-control25 patients
25 HC
Plasma[77]
let-7a-5p, let-7b-5p, let-7d-5p, let-7f-5p, let-7g-5p, let-7i-5p, 199a-3p, 320a-3p, 320b, 320c, 320d, 320e, 328-3p, 331-3pTrizol reagentqRT-PCRCross sectional19 OE patients
21 HC
Plasma[96]
200c-3pTRIzol reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional27 endometriosis patients, 12 uterine leiomyoma or hysterectomy in patients with grade II–III cervical intraepithelial neoplasiaEctopic endometrial tissues[76]
9-5p, 34b-3p, 34c-5pTrizol reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional4 patients
3 HC
Endometrial biopsies[97]
Chronic secondary musculoskeletal painlet-7d-5p, 98-5p
29c-3p, 205-5p, 222-3p, 320a-3p, 423-5p
↑ (No vs Np)
↓ (No vs Np)
Exiqon miRCURY RNA isolation (Exiqon, Vedbaek, Denmark)qRT-PCRCross-sectional100 patients
(60 No, 40 Np)
Plasma[79]
210Isothiocyanatephenol/chloroform extractionqRT-PCRCross-sectional20 early-stage OA patients, 20 late-stage OA patients
10 HC
Synovial fluid from knee joints[83]
146a-5pFor serum: miRCURY RNA Isolation Kit–Biofluids (Exiqon, Denmark);
for cartilage: miRVana miRNA isolation kit (Applied Biosysstems, Life Technologies, Carlsbad, CA, USA)
qRT-PCRCross-sectional28 OA patients
2 HC
Cartilage tissue and serum[84]
34a-5pTRIzol reagent (Invitrogen, Carlsbad, CA, USA)qRT-PCRCross-sectional15 OA patients
10 HC
Cartilage tissue and cultured primary chondrocytes[85]
665RNeasy Mini Kit (Qiagen, Valencia, CA, USA)qRT-PCRCross-sectional30 OA patients
5 HC
Cartilage tissue and cultured primary chondrocytes[88]
let-7e-5p, 454-5p
885-5p

miRNeasy kit (Qiagen, Inc., Valencia, CA, USA)qRT-PCRProspective cohort (Bruneck study)entire cohort (816 subjects) Serum[98]
16-5p, 29c-3p, 93-5p, 126-3p, 146a-5p, 184, 186-5p, 195-5p, 345-5p, 885-5pmiRNeasy kit (Qiagen, Inc., Valencia, CA, USA)qRT-PCRCross-sectional27 OA patients
27 HC
Plasma [99]
9-5p, 138-5p, 146a-5p, 335-5pmirVana miRNA Isolation Kit (Exiqon, Vedbaek, Denmark)qRT-PCRCross-sectional40 early and late OA patients
2 HC
Cartilage[100]
146a-5p, 155-5p, 181a-5p, 223-3p Trizol reagent (Invitrogen, Carlsbad, CA, USA) qRT-PCRCross-sectional36 OA patients
36 HC
PBMC[100]
23a-3p, 30c-5pmiRNeasy Serum/Plasma Kit (Qiagen, Germany)qRT-PCRProspective (before and 6 months after high tibial osteotomy)22 patients with medial compartmental knee OASynovial fluid[89]
Legend: BPS: bladder pain syndrome; CNP: chronic neuropathic pain; HC: healthy controls; IC: Interstitial cystitis; IDD: Intervertebral disc degeneration; No: nociceptive pain; Np: neuropathic pain; OE: ovarian endometriosis; OA: osteoarthritis; NP: nucleus pulposus; PBMC: peripheral blood mononuclear cell; qRT-PCR: Quantitative real-time PCR; TLDA: TaqMan Low Density Array. ↓/↑, difference in miRNAs expression, respectively up- and down-expressed.

2.3. Bioinformatic Analysis of miRNA Gene Targets and Pathways

Using the miRNAs found in at least 3 studies, i.e., for CPP: let-7a-5p, hsa-miR-34a-5p, hsa-miR-103a-3p, hsa-miR-155-5p; for CSP: hsa-miR-146a-5p, hsa-miR-185-5p, hsa-miR-320a-3p; we obtained the following targets: let-7a-5p: 632 targets; hsa-miR-34a-5p: 419 targets; hsa-miR-103a-3p: 388 targets; hsa-miR-155-5p: 145 targets; 146a-5p: 58 targets; hsa-miR-185-5p: 165 targets; hsa-miR-320a-3p: 377 targets. As created in Cytoscape, the network consists of 1866 nodes and 2183 edges (Figure 1). Detailed information about the miRNA-gene target is listed in Supplementary Materials (Table S1). Considering that mRNAs are typically targeted by many miRNAs and that each miRNA targets multiple mRNAs, the miRNA-gene pairs identified are part of co-regulation and interaction networks where different mRNA targets overlap between different miRNAs and between CPP and CSP.
Then, we performed a KEGG pathway analysis to identify the possible signaling pathways related to CPP and CSP. The analysis was performed by separating the two types of pain in order to potentially discriminate between CPP and CSP. Therefore, for a more robust and convincing analysis, targets of all miRNAs were taken together, and duplicates were removed. In that way, 1438 overlapping targets for CPP and 566 overlapping targets for CSP are identified and analyzed with GeneTrail2.
We identified in total 131 and 77 KEGG pathways for CPP and CSP, respectively. A deeper insight into these pathways showed that the vast majority is associated with cellular processes. In particular, for CPP we found pathways involved in signal transduction such as PI3K-Akt signaling, MAPK signaling pathway, RAP1 signaling pathway and Ras signaling pathway; in the immune system, such as Fc gamma R-mediated phagocytosis, Leukocyte transendothelial migration and a Chemokine signaling pathway and in the endocrine system, such as melanogenesis and the insulin signaling pathway (Figure 2). Additionally, for CSP we found pathways involved in signal transduction, such as the ErbB signaling pathway, the mTor signaling pathway, MAPK signaling pathways, PI3K-Akt signaling pathways and the Rap1 signaling pathway; in the immune system, such as the Toll-like receptor signaling pathway, the Chemokine signaling pathway and in the endocrine system, such as the Insulin signaling pathway (Figure 3). Interestingly, some of the main pathways for CPP are also related to the function of the nervous system, such as the Neurotrophin signaling pathway, Dopaminergic synapse and Cholinergic synapse, and to substance dependence, such as amphetamine addiction, morphine addiction and cocaine addiction (Figure 2). For both CPP and CSP some other pathways are associated with diseases such as pathways in cancer and viral infectious diseases (Figure 4 and Figure 5). See Supplementary Material for further details (Tables S2 and S3).

3. Discussion

To the best of our knowledge, this is the first review to summarize the human studies on miRNA expression associated with CPP and CSP conditions, and the first to highlight the potential biological mechanisms leading to pathological conditions. After peripheral inflammation or nerve injury, the increase in inflammatory mediators causes a change in the expression of some miRNAs, resulting in an alteration in pain-related genes. Such an alteration leads to an increase in dorsal root ganglion neuronal excitability, spinal central sensitization and hyperalgesia and allodynia, hence the chronic pain.
The data presented here indicate that the dysregulation of some miRNAs is associated with the pain condition, suggesting their potential role in the pathogenesis of certain diseases and the pain process. For example, miR-34a-5p, already known for its role in regulating the inflammatory response and vascular endothelial stress response [101], could be considered as a potential candidate in the pathogenesis of migraines [38,39,54]. It is involved in pathways regulating the function of nervous system, such as the Dopaminergic synapse and GABAergic signaling. Andersen et al. found that this miRNA negatively modulated the expression of genes that facilitate GABAergic signaling, suggesting a facilitator transmission that promotes the nociceptive effect [54]. Moreover, miR-34a-5p could be considered as biomarker for therapeutic response prediction. It is known that drug administration can indirectly affect miRNA expression [102], and some specific miRNAs could be relevant as indicators of a drug response [103]. A decreased expression of miR-34a-5p was found in the study of Gallelli et al., in patients under drug treatment compared to untreated subjects [38]. A growing body of evidence indicates that miRNAs can be useful in monitoring therapeutic responses, and knowledge of the involvement of miRNAs in the induction and progression of CP may open the opportunity to develop miRNA-based therapeutics as a new analgesic modality. Of course, there are still many challenges, including the rational selection of miRNAs to target, the delivery system and the route of administration (systemic vs tissue), as well as the stage of painful disease in which targeted miRNAs play a role. Another prominent miRNA featured in this review was miR-103a-3p, belonging to the so-called miR-15/107 group of miRNA genes involved in the regulation of metabolism, cell division, stress responses and exercise [104]. This miRNA was mainly found in studies conducted in patients with FM [24,26,50] and is involved in different pathways such as the SNRK/NF-κB/p65 signaling pathway [105]. An elevated gene expression of miR-103a-3p can lead to the suppression of SNRK and to an over-activation of the pro-inflammatory transcription factor NF-κB. This machinery is linked to mechanisms promoting epigenetic resilience and strategies to limit damage due to diseases [106], as well as synaptic plasticity, which results in adaptive processes [107]. MiR-155-5p has here emerged as being associated with CPP, in particular with TN, migraines and musculoskeletal pain. Its expression is induced by factors associated with infections, injury, hypoxia and inflammation, through signaling pathways involving NF-κB activation; contrarily, it is decreased by anti-inflammatory cytokines, resolvins and glucocorticoids, thus providing a feedback inhibition of the miR-155-induced responses [108]. MiR-155-5p plays a multifunctional role in innate and adaptive immunity and inflammation, so that its aberrant overexpression represents a potential disease biomarker or therapeutic target in states of chronic inflammation [109]. Its involvement in neuroinflammation, vascular dysfunction and neurodegeneration has been documented in animal and human studies [110,111]. Interestingly, a miR-155-5p target, i.e., NRG3 (neuregulin-3), is a ligand of ERBB4, and is thought to regulate neuroblast proliferation, migration and differentiation; repair after nerve injury; synapse formation; regulation of glutamate; GABA and dopamine release [112]. Recent studies point to an important role for miR-155-5p in the development of neuropathic pain and associated mechanical allodynia, thermal hyperalgesia and neuronal inflammation, through mechanisms involving the regulation of the suppressor of cytokine signalling 1 (SOCS1) and TNF-α receptor-transient receptor potential ankyrin 1 (TNFR1-TRPA1) pathways [113,114,115].
Another miRNA related to CPP is let-7a-5p, a highly evolutionarily conserved miRNA that resulted in downregulation in patients with FM and CRPS. Let-7a-5p has been reported to regulate stem-cell differentiation, neuromuscular development and cell proliferation and differentiation, and, like other members of the let-7 family, it seems to function as a tumor suppressor [116,117]. The upregulation of let-7a-5p in microglia has been found to correlate with protection from ischemia and neuroinflammation by inhibiting the expression of pro-inflammatory factors (e.g., IL-6, iNOS) and boosting the expression of anti-inflammatory factors (IL-10 and IL-4) [118]. Interestingly, in cellular and animal models, chronic treatment with morphine led to an increase in brain let-7 miRNAs expression and to a parallel decrease in the expression of μ-opioid receptor, thus implicating let-7 in the mechanism of opioid tolerance [119], which limits the analgesic efficacy of opioids.
A downregulation of miR-185-5p was observed in CSP conditions including IDD, chronic secondary visceral pain and post-traumatic pain. MiR-185-5p has been found to protect against excessive extracellular matrix degradation associated with inflammation and degeneration in human IDD samples: indeed, here, the higher expression of a circular RNA derived from tissue inhibitor of metallopeptidases 2 (circ-TIMP2) led to the sequestration of miR-185-5p and the consequent upregulation of miR-185-5p target genes, including MMP-2, associated with extracellular matrix catabolism [95]. These data are in agreement with other animal findings in which miR-185-5p suppressed autophagy and apoptosis of nucleus pulposus cells in IDD via inhibition of the Wnt/β-catenin signaling pathway [120]. Furthermore, anti-inflammatory effects have been observed for miR-185-5p in neuropathic pain induced by experimental chronic constrictive injury, where miR-185-5p was reduced in spinal cord tissues and its upregulation attenuated mechanical and thermal hyperalgesia and decreased microglia and astrocytes recruitment and the levels of inflammatory cytokines by repressing NLRP3 inflammasome [121].
Another pain-associated miRNA is miR-320a-3p, which has here emerged as mostly associated with CSP; an important finding from the study by Dayer et al. was its predictive value, in combination with miR-98-5p, for the discrimination between the nociceptive and neuropathic origin of pain [79], so that it resulted in downregulation and upregulation, respectively. Similarly, its expression is deregulated in secondary visceral pain conditions, as well as in post-traumatic pain. Downregulation of miR-320a-3p might lead to heightened inflammation through the upregulation of its target’s MAPK1 signaling pathway [122] or the E2F family of transcription factors, which have been linked to neuronal death, neuroinflammation and tissue damage [123]. Several miR-320a-3p targets are related to stress response and pain, including NR3C1 and FKBP5, and are involved in chronic musculoskeletal pain after traumatic events [60].
Additionally, miR-146a-5p has been found to be consistently associated with OA [57,84,89,100,124] in different tissues such as plasma, cartilage and blood cells, thus reflecting intra- and extracellular events and representing a potential candidate systemic biomarker of the processes associated with the disease. Its expression is induced in response to an increase in the levels of inflammatory cytokines, such as tumor necrosis factor α and interleukin (IL)-1β, and to alteration of inflammatory factors such as the Toll-like receptor (TLR) and NF-κB. These processes constitute a negative feedback loop that controls immune activation and may modulate OA-related inflammation in early-stage OA through the miR-146a-5p-mediated downregulation of adaptor/scaffold proteins such as IRAK1 and TRAF6 involved in pro-inflammatory and catabolic pathways, including the IL-1 and TLR signaling pathway, known to positively regulate NF-κB [125]. In later stages, miR-146a-5p levels become low in parallel with degenerative changes and progressive degradation of cartilage [100,126], which is associated with pain. In an animal model of OA-related knee joint pain, a dysregulated expression of miR-146a-5p was also found in sensory neurons of the peripheral and central nervous system, thus suggesting its involvement in both the peripheral (knee) and central pain transmission; moreover, its upregulation in glial cells decreased the expression of pain-related inflammatory cytokines and pain mediators (TNFα, COX-2, iNOS, IL-6, IL-8, RANTS and ion channel, TRPV1) [125].
Many pain conditions, however, remain insufficiently characterized in terms of miRNA expression changes or have not been associated with specific miRNAs, thus requiring further studies to better characterize the molecular (miRNA) fingerprint of pain. For instance, a variety of miRNAs have been intensively researched and investigated in cancers, but the involvement of miRNAs in cancer-related pain is poorly understood.
The bioinformatic analysis of the dysregulated miRNAs showed that the predicted miRNA targets play a role in several cellular functions and cell signaling. The most relevant pathways are involved in signal transduction (e.g., PI3K-Akt signaling, MAPK signaling, Ras signaling, ErbB signaling, Calcium signaling), the endocrine system (e.g., insulin signaling), the immune system (e.g., Toll-like receptor signaling, Leukocyte transendothelial migration, Chemokine signaling) and neuronal functions (e.g., neurotrophin signaling, glutamatergic synapse, GABAergic synapse, cholinergic synapse) that have been associated with pain pathology [127]. For instance, the intracellular signal transduction pathways found here to be associated with miRNA targets are known to play critical roles in pain processes involving neurotransmission, inflammation, neuronal plasticity and hypersensitivity through the modification of receptor/channel phosphorylation or the long-term adaptive regulation of gene transcription and translation [128]. Similarly, tissue infiltration and activation of immune cells at the site of injury, including mast cells, neutrophils, macrophages and T lymphocytes, as well as glial cells, and their release of inflammatory factors, mainly TNF- α, interleukins, interferon- α, prostaglandins, chemokines and leukocyte elastase, are involved in the development and progression of pain [129,130].
A great advancement in pain research and clinical application would be the possibility of discriminating between primary and secondary pain. Besides, though very preliminary, the observation that some miRNAs and related pathways are differentially deregulated in primary and secondary CP would pave the way for confirmatory experimental studies on the potential use of miRNAs for differential diagnosis between these two painful conditions. Our computational analysis suggests that although nearly the same pathways are predicted targets by miRNAs in CPP and CSP, a greater pathways enrichment can be observed in CPP compared with CSP. Moreover, in CPP, several pathways related to neuronal function have also been identified, and most of them are implicated in the transmission and regulation of pain, such as the Neurotrophin signaling pathway [131]; synapse function, e.g., Dopaminergic synapse [132]; Cholinergic synapse; Glutamatergic synapse; Serotonergic synapse; GABAergic synapse [20] and synapse plasticity (e.g., long-term potentiation). Interestingly, pathways related to substance dependence (e.g., amphetamine addiction, morphine addiction, cocaine addiction, alcoholism, nicotine addiction) also emerge in CPP, in accordance with previous propositions that there are anatomical, clinical and neurobiological similarities between chronic pain and addiction, which include overlapping neural circuitry and neuroadaptations including reward deficiency, impaired inhibitory control, compulsive drug seeking and high stress [133,134]. Therefore, miRNAs may represent a proximal cue in the biopsychosocial system of chronic pain and a biological link between chronic pain and neurobiological dependence that deserves further investigations.
This review highlights the use of diversified methodologies in miRNA analysis. Some factors, such as the heterogeneity in the tissue samples and the different extraction and detection techniques, make the comparison among studies hard, suggesting the need for a standardized approach.
The analyzed studies have some limitations. Considering the frequent low differences between miRNA expression in diseased versus healthy subjects, it is important to control some factors such as miRNA pre-processing and normalization experiments, data processing and optimization. In this regard, qRT-PCR is the most widely used method, which can be considered the “gold standard” for the verification of microarrays and next-generation sequencing results due to its greater precision and sensitivity, rapidity and ease of use [135]. In addition, the small number of participants and the lack of result validation in independent cohorts could lead to less coherent results, which is why the works with a limited sample size and those with poor study design should be considered with caution. An important aspect to consider is that, in most studies concerning miRNA profile in CSP, comparisons were made between healthy controls and painful diseased subjects, thus limiting the ability to discern whether miRNAs are pain-specific or are mainly associated with—or modulated by—the disease causing pain. Again, the lacking or insufficient information on gender differences or the impact of age, BMI and medications (including analgesic treatments), which can affect miRNA expression, can also lead to heterogeneity of study results. In many studies analyzed here, no data were reported on the association of deregulated miRNAs with pain symptoms and their evolution in the cohorts studied. Therefore, further longitudinal prospective studies should be conducted because miRNA regulation can change over time during the course of painful disease, and other pathological conditions or confounding factors can overlap over time, which can impact miRNA levels. Importantly, a high throughput analysis of miRNAs was not conducted in the available studies, and only a set of miRNAs or often individual miRNAs were selected for the analysis, so the possibility of missing important miRNAs as potential biomarkers of CP could not be excluded. Information on the role or function of most of miRNA candidates remains incomplete, and, therefore, involvement in the induction and/or maintenance of CP is unknown. Indeed, most studies adopted in silico predictive analysis, so further experimental in vitro or in vivo studies are warranted to validate the targets and determine if the predicted miRNA/mRNA interaction results in significant biological changes.
Notwithstanding limitations and challenges, miRNA research has grown to improve the understanding of the potential role for these molecules in pain physiopathology, eventually enabling this role to be exploited for pain monitoring and management.

4. Materials and Methods

4.1. Literature Search Strategy

An electronic literature search of MEDLINE/Pubmed, ISI Web of Knowledge, Scopus and Google Scholar was conducted by three separate investigators to retrieve relevant studies concerning the expression of miRNAs in the pathogenesis of CPP and CSP conditions. The following keywords were used: chronic pain, pain syndrome, painful disorders, miRNA, miR, microRNA, pain diagnosis, pain biomarker, pain pathophysiology, chronic primary pain, chronic secondary pain, in conjunction with keywords related to the different CPP and CSP conditions, as specified in Table 1. Additionally, references from selected original studies and reviews were scrutinized for further relevant evidence. The following inclusion criteria guided the search and selection of studies: all types of study design; publication until July 2021; English language; human studies. We excluded studies that used only a microarray for miRNA detection, since, compared to the other molecular approaches, it may give less reliable results due to its low detection sensitivity, reproducibility and dynamic range [136].

4.2. Bioinformatic Analysis

To better understand the potential pathophysiological impact of miRNAs, an in silico analysis was performed to reveal their target genes. For the bioinformatic analysis, dysregulated miRNAs were selected among those reported as implicated in CP by at least three independent studies to ensure consistency. For a more robust selection of the potential target genes, we adopted three widely used databases, i.e., TargetScanVert and miRDB, accessed through miRbase [21] and miRWalk, a resource hosting the predicted as well as the experimentally validated miRNA-target interactions [137]. To further improve the reliability of these results, InteractiVenn [138] was used as a tool to retrieve the overlapping target genes, which were subsequently used for pathway enrichment analyses. Cytoscape software (version 3.7.1) was used to construct the miRNA–mRNA regulatory network of miRNA and their target genes [139].
Pathway enrichment analyses were conducted to better understand the functional role of dysregulated miRNAs. GeneTrail2 was used (version 1.6) [140] as a platform to access the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, using the following settings: over-representation analysis; two-sided as null hypothesis (for p-value computation); Benjamini-Yekutieli as method to adjust p-values; 0.05 as significance level.
Figures were prepared using R packages [141].

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms23116016/s1.

Author Contributions

Conceptualization, E.S. and M.R.T.; methodology, A.P., E.S. and M.R.T.; literature search, all the authors; writing—original draft preparation, A.P., E.S. and M.R.T.; writing—review and editing, all the authors; supervision S.S., P.M., C.G.L., S.G., T.G., F.B., A.D.D., E.S. and M.R.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Raja, S.N.; Carr, D.B.; Cohen, M.; Finnerup, N.B.; Flor, H.; Gibson, S.; Keefe, F.J.; Mogil, J.S.; Ringkamp, M.; Sluka, K.A.; et al. The revised International Association for the Study of Pain definition of pain: Concepts, challenges, and compromises. Pain 2020, 161, 1976–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Scholz, J.; Finnerup, N.B.; Attal, N.; Aziz, Q.; Baron, R.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Cruccu, G.; Davis, K.D.; et al. The IASP classification of chronic pain for ICD-11: Chronic neuropathic pain. Pain 2019, 160, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Treede, R.-D.; Rief, W.; Barke, A.; Aziz, Q.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Evers, S.; Finnerup, N.B.; First, M.B.; et al. Chronic pain as a symptom or a disease: The IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain 2019, 160, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Breivik, H.; Collett, B.; Ventafridda, V.; Cohen, R.; Gallacher, D. Survey of chronic pain in Europe: Prevalence, impact on daily life, and treatment. Eur. J. Pain 2006, 10, 287. [Google Scholar] [CrossRef]
  5. Dahlhamer, J.; Lucas, J.; Zelaya, C.; Nahin, R.; Mackey, S.; DeBar, L.; Kerns, R.; Von Korff, M.; Porter, L.; Helmick, C. Prevalence of Chronic Pain and High-Impact Chronic Pain Among Adults—United States, 2016. MMWR Morb. Mortal. Wkly. Rep. 2018, 67, 1001–1006. [Google Scholar] [CrossRef]
  6. Abbafati, C.; Abbas, K.M.; Abbasi-Kangevari, M.; Abd-Allah, F.; Abdelalim, A.; Abdollahi, M.; Abdollahpour, I.; Abegaz, K.H.; Abolhassani, H.; Aboyans, V.; et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
  7. Bernfort, L.; Gerdle, B.; Rahmqvist, M.; Husberg, M.; Levin, L. Severity of chronic pain in an elderly population in Sweden—Impact on costs and quality of life. Pain 2015, 156, 521–527. [Google Scholar] [CrossRef] [Green Version]
  8. Azevedo, L.F.; Costa-Pereira, A.; Mendonça, L.; Dias, C.C.; Castro-Lopes, J. The economic impact of chronic pain: A nationwide population-based cost-of-illness study in Portugal. Eur. J. Health Econ. 2014, 17, 87–98. [Google Scholar] [CrossRef]
  9. Cohen, S.P.; Vase, L.; Hooten, W.M. Chronic pain: An update on burden, best practices, and new advances. Lancet 2021, 397, 2082–2097. [Google Scholar] [CrossRef]
  10. Balzani, E.; Fanelli, A.; Malafoglia, V.; Tenti, M.; Ilari, S.; Corraro, A.; Muscoli, C.; Raffaeli, W. A Review of the Clinical and Therapeutic Implications of Neuropathic Pain. Biomedicines 2021, 9, 1239. [Google Scholar] [CrossRef]
  11. Huang, Y.; Gu, M.; Tang, Y.; Sun, Z.; Luo, J.; Li, Z. Systematic review and meta-analysis of prognostic microRNA biomarkers for survival outcome in laryngeal squamous cell cancer. Cancer Cell Int. 2021, 21, 316. [Google Scholar] [CrossRef] [PubMed]
  12. Leo, C.G.; Mincarone, P.; Tumolo, M.R.; Panico, A.; Guido, M.; Zizza, A.; Guarino, R.; De Santis, G.; Sedile, R.; Sabina, S. MiRNA expression profiling in HIV pathogenesis, disease progression and response to treatment: A systematic review. Epigenomics 2021, 13, 1653–1671. [Google Scholar] [CrossRef] [PubMed]
  13. Catalanotto, C.; Cogoni, C.; Zardo, G. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. Int. J. Mol. Sci. 2016, 17, 1712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tüfekci, K.U.; Meuwissen, R.L.J.; Genç, Ş. The Role of MicroRNAs in Biological Processes. In Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2014; Volume 1107, pp. 15–31. [Google Scholar] [CrossRef]
  15. Machado, M.T.; Navega, S.; Dias, F.; de Sousa, M.J.C.; Teixeira, A.L.; Medeiros, R. microRNAs for peripheral blood fraction identification: Origin, pathways and forensic relevance. Life Sci. 2015, 143, 98–104. [Google Scholar] [CrossRef]
  16. Hanson, E.K.; Lubenow, H.; Ballantyne, J. Identification of forensically relevant body fluids using a panel of differentially expressed microRNAs. Anal. Biochem. 2009, 387, 303–314. [Google Scholar] [CrossRef]
  17. Turchinovich, A.; Weiz, L.; Burwinkel, B. Extracellular miRNAs: The mystery of their origin and function. Trends Biochem. Sci. 2012, 37, 460–465. [Google Scholar] [CrossRef]
  18. Polli, A.; Godderis, L.; Ghosh, M.; Ickmans, K.; Nijs, J. Epigenetic and miRNA Expression Changes in People with Pain: A Systematic Review. J. Pain 2020, 21, 763–780. [Google Scholar] [CrossRef]
  19. Riolo, G.; Cantara, S.; Marzocchi, C.; Ricci, C. miRNA Targets: From Prediction Tools to Experimental Validation. Methods Protoc. 2020, 4, 1. [Google Scholar] [CrossRef]
  20. Dinakar, P.; Stillman, A.M. Pathogenesis of Pain. Semin. Pediatr. Neurol. 2016, 23, 201–208. [Google Scholar] [CrossRef]
  21. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef]
  22. Nicholas, M.; Vlaeyen, J.W.; Rief, W.; Barke, A.; Aziz, Q.; Benoliel, R.; Cohen, M.; Evers, S.; Giamberardino, M.A.; Goebel, A.; et al. The IASP classification of chronic pain for ICD-11: Chronic primary pain. Pain 2019, 160, 28–37. [Google Scholar] [CrossRef] [PubMed]
  23. Wolfe, F.; Clauw, D.J.; Fitzcharles, M.-A.; Goldenberg, D.L.; Katz, R.S.; Mease, P.; Russell, A.S.; Russell, I.J.; Winfield, J.B.; Yunus, M.B. The American College of Rheumatology Preliminary Diagnostic Criteria for Fibromyalgia and Measurement of Symptom Severity. Arthritis Care Res. 2010, 62, 600–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Braun, A.; Evdokimov, D.; Frank, J.; Sommer, C.; Üçeyler, N. MiR103a-3p and miR107 are related to adaptive coping in a cluster of fibromyalgia patients. PLoS ONE 2020, 15, e0239286. [Google Scholar] [CrossRef] [PubMed]
  25. Masotti, A.; Baldassarre, A.; Guzzo, M.P.; Iannuccelli, C.; Barbato, C.; Di Franco, M. Circulating microRNA Profiles as Liquid Biopsies for the Characterization and Diagnosis of Fibromyalgia Syndrome. Mol. Neurobiol. 2016, 54, 7129–7136. [Google Scholar] [CrossRef] [PubMed]
  26. Leinders, M.; Doppler, K.; Klein, T.; Deckart, M.; Rittner, H.; Sommer, C.; Üçeyler, N. Increased cutaneous miR-let-7d expression correlates with small nerve fiber pathology in patients with fibromyalgia syndrome. Pain 2016, 157, 2493–2503. [Google Scholar] [CrossRef]
  27. Philippou, A.; Halapas, A.; Maridaki, M.; Koutsilieris, M. Type I insulin-like growth factor receptor signaling in skeletal muscle regeneration and hypertrophy. J. Musculoskelet. Neuronal Interact. 2007, 7, 208–218. [Google Scholar]
  28. Orlova, I.A.; Alexander, G.M.; Qureshi, R.A.; Sacan, A.; Graziano, A.; Barrett, J.E.; Schwartzman, R.J.; Ajit, S.K. MicroRNA modulation in complex regional pain syndrome. J. Transl. Med. 2011, 9, 195. [Google Scholar] [CrossRef] [Green Version]
  29. Douglas, S.R.; Shenoda, B.B.; Qureshi, R.A.; Sacan, A.; Alexander, G.M.; Perreault, M.; Barrett, J.E.; Aradillas-Lopez, E.; Schwartzman, R.J.; Ajit, S.K. Analgesic Response to Intravenous Ketamine Is Linked to a Circulating microRNA Signature in Female Patients with Complex Regional Pain Syndrome. J. Pain 2015, 16, 814–824. [Google Scholar] [CrossRef]
  30. Shenoda, B.B.; Tian, Y.; Alexander, G.M.; Aradillas-Lopez, E.; Schwartzman, R.J.; Ajit, S.K. miR-34a-mediated regulation of XIST in female cells under inflammation. J. Pain Res. 2018, 11, 935–945. [Google Scholar] [CrossRef] [Green Version]
  31. Ramanathan, S.; Douglas, S.R.; Alexander, G.M.; Shenoda, B.B.; Barrett, J.E.; Aradillas, E.; Sacan, A.; Ajit, S.K. Exosome microRNA signatures in patients with complex regional pain syndrome undergoing plasma exchange. J. Transl. Med. 2019, 17, 81. [Google Scholar] [CrossRef]
  32. Yadav, Y.R.; Nishtha, Y.; Sonjjay, P.; Vijay, P.; Shailendra, R.; Yatin, K. Trigeminal neuralgia. Asian J. Neurosurg. 2017, 12, 585–597. [Google Scholar] [CrossRef] [PubMed]
  33. Li, X.; Wang, D.; Zhou, J.; Yan, Y.; Chen, L. Evaluation of circulating microRNA expression in patients with trigeminal neuralgia: An observational study. Medicine 2020, 99, e22972. [Google Scholar] [CrossRef] [PubMed]
  34. Pietrobon, D.; Moskowitz, M.A. Pathophysiology of Migraine. Annu. Rev. Physiol. 2013, 75, 365–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Cheng, C.-Y.; Chen, S.-P.; Liao, Y.-C.; Fuh, J.-L.; Wang, Y.-F.; Wang, S.-J. Elevated circulating endothelial-specific microRNAs in migraine patients: A pilot study. Cephalalgia 2017, 38, 1585–1591. [Google Scholar] [CrossRef] [PubMed]
  36. Weber, C.; Schober, A.; Zernecke, A. MicroRNAs in arterial remodelling, inflammation and atherosclerosis. Curr. Drug Targets 2010, 11, 950–956. [Google Scholar] [CrossRef]
  37. Urbich, C.; Kuehbacher, A.; Dimmeler, S. Role of microRNAs in vascular diseases, inflammation, and angiogenesis. Cardiovasc. Res. 2008, 79, 581–588. [Google Scholar] [CrossRef] [Green Version]
  38. Gallelli, L.; Cione, E.; Peltrone, F.; Siviglia, S.; Verano, A.; Chirchiglia, D.; Zampogna, S.; Guidetti, V.; Sammartino, L.; Montana, A.; et al. Hsa-miR-34a-5p and hsa-miR-375 as Biomarkers for Monitoring the Effects of Drug Treatment for Migraine Pain in Children and Adolescents: A Pilot Study. J. Clin. Med. 2019, 8, 928. [Google Scholar] [CrossRef] [Green Version]
  39. Greco, R.; De Icco, R.; DeMartini, C.; Zanaboni, A.M.; Tumelero, E.; Sances, G.; Allena, M.; Tassorelli, C. Plasma levels of CGRP and expression of specific microRNAs in blood cells of episodic and chronic migraine subjects: Towards the identification of a panel of peripheral biomarkers of migraine? J. Headache Pain 2020, 21, 122. [Google Scholar] [CrossRef]
  40. Saha, L. Irritable bowel syndrome: Pathogenesis, diagnosis, treatment, and evidence-based medicine. World J. Gastroenterol. 2014, 20, 6759–6773. [Google Scholar] [CrossRef]
  41. Zhou, Q.; Yang, L.; Larson, S.; Basra, S.; Merwat, S.; Tan, A.; Croce, C.; Verne, G.N. Decreased miR-199 augments visceral pain in patients with IBS through translational upregulation of TRPV1. Gut 2015, 65, 797–805. [Google Scholar] [CrossRef]
  42. Dhaka, A.; Uzzell, V.; Dubin, A.E.; Mathur, J.; Petrus, M.J.; Bandell, M.; Patapoutian, A. TRPV1 Is Activated by Both Acidic and Basic pH. J. Neurosci. 2009, 29, 153–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zhu, H.; Xiao, X.; Shi, Y.; Wu, Y.; Huang, Y.; Li, D.; Xiong, F.; He, G.; Chai, Y.; Tang, H. Inhibition of miRNA-29a regulates intestinal barrier function in diarrhea-predominant irritable bowel syndrome by upregulating ZO-1 and CLDN1. Exp. Ther. Med. 2020, 20, 155. [Google Scholar] [CrossRef] [PubMed]
  44. Mahurkar-Joshi, S.; Rankin, C.R.; Videlock, E.J.; Soroosh, A.; Verma, A.; Khandadash, A.; Iliopoulos, D.; Pothoulakis, C.; Mayer, E.A.; Chang, L. The Colonic Mucosal MicroRNAs, MicroRNA-219a-5p, and MicroRNA-338-3p Are Downregulated in Irritable Bowel Syndrome and Are Associated with Barrier Function and MAPK Signaling. Gastroenterology 2021, 160, 2409–2422.e19. [Google Scholar] [CrossRef] [PubMed]
  45. Airaksinen, O.; Brox, J.I.; Cedraschi, C.; Hildebrandt, J.; Klaber-Moffett, J.; Kovacs, F.; Mannion, A.F.; Reis, S.; Staal, J.B.; Ursin, H.; et al. Chapter 4 European guidelines for the management of chronic nonspecific low back pain. Eur. Spine J. 2006, 15, s192–s300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Luchting, B.; Heyn, J.; Hinske, L.C.; Azad, S.C. Expression of miRNA-124a in CD4 Cells Reflects Response to a Multidisciplinary Treatment Program in Patients with Chronic Low Back Pain. Spine 2017, 42, E226–E233. [Google Scholar] [CrossRef]
  47. Uchida, S.; Hara, K.; Kobayashi, A.; Funato, H.; Hobara, T.; Otsuki, K.; Yamagata, H.; McEwen, B.S.; Watanabe, Y. Early Life Stress Enhances Behavioral Vulnerability to Stress through the Activation of REST4-Mediated Gene Transcription in the Medial Prefrontal Cortex of Rodents. J. Neurosci. 2010, 30, 15007–15018. [Google Scholar] [CrossRef]
  48. Dwivedi, Y. Emerging role of microRNAs in major depressive disorder: Diagnosis and therapeutic implications. Dialog. Clin. Neurosci. 2014, 16, 43–61. [Google Scholar] [CrossRef]
  49. Bjersing, J.L.; Lundborg, C.; Bokarewa, M.I.; Mannerkorpi, K. Profile of Cerebrospinal microRNAs in Fibromyalgia. PLoS ONE 2013, 8, e78762. [Google Scholar] [CrossRef] [Green Version]
  50. Bjersing, J.L.; Bokarewa, M.I.; Mannerkorpi, K. Profile of circulating microRNAs in fibromyalgia and their relation to symptom severity: An exploratory study. Rheumatol. Int. 2014, 35, 635–642. [Google Scholar] [CrossRef]
  51. Cerdá-Olmedo, G.; Mena-Durán, A.V.; Monsalve, V.; Oltra, E. Identification of a MicroRNA Signature for the Diagnosis of Fibromyalgia. PLoS ONE 2015, 10, e0121903. [Google Scholar] [CrossRef] [Green Version]
  52. Tafuri, E.; Santovito, D.; De Nardis, V.; Marcantonio, P.; Paganelli, C.; Affaitati, G.; Bucci, M.; Mezzetti, A.; Giamberardino, M.A.; Cipollone, F. MicroRNA profiling in migraine without aura: Pilot study. Ann. Med. 2015, 47, 468–473. [Google Scholar] [CrossRef] [PubMed]
  53. Zhai, Y.; Zhu, Y.-Y. MiR-30a relieves migraine by degrading CALCA. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2022–2028. [Google Scholar] [PubMed]
  54. Andersen, H.H.; Duroux, M.; Gazerani, P. Serum MicroRNA Signatures in Migraineurs During Attacks and in Pain-Free Periods. Mol. Neurobiol. 2015, 53, 1494–1500. [Google Scholar] [CrossRef] [PubMed]
  55. Liao, X.-J.; Mao, W.-M.; Wang, Q.; Yang, G.-G.; Wu, W.-J.; Shao, S.-X. MicroRNA-24 inhibits serotonin reuptake transporter expression and aggravates irritable bowel syndrome. Biochem. Biophys. Res. Commun. 2016, 469, 288–293. [Google Scholar] [CrossRef]
  56. Brat, G.A.; Agniel, D.; Beam, A.; Yorkgitis, B.; Bicket, M.; Homer, M.; Fox, K.P.; Knecht, D.B.; McMahill-Walraven, C.N.; Palmer, N.; et al. Postsurgical prescriptions for opioid naive patients and association with overdose and misuse: Retrospective cohort study. BMJ 2018, 360, j5790. [Google Scholar] [CrossRef] [Green Version]
  57. Giordano, R.; Petersen, K.K.; Andersen, H.H.; Lichota, J.; Valeriani, M.; Simonsen, O.; Arendt-Nielsen, L. Preoperative serum circulating microRNAs as potential biomarkers for chronic postoperative pain after total knee replacement. Mol. Pain 2020, 16. [Google Scholar] [CrossRef]
  58. Vucetic, M.; Roganovic, J.; Freilich, M.; Shafer, D.; Milic, M.; Djukic, L.; Petrovic, N.; Markovic, E.; Markovic, A.; Brkovic, B. Bone microRNA-21 as surgical stress parameter is associated with third molar postoperative discomfort. Clin. Oral Investig. 2020, 25, 319–328. [Google Scholar] [CrossRef]
  59. Linnstaedt, S.D.; Walker, M.G.; Parker, J.S.; Yeh, E.; Sons, R.L.; Zimny, E.; Lewandowski, C.; Hendry, P.L.; Damiron, K.; Pearson, C.; et al. MicroRNA circulating in the early aftermath of motor vehicle collision predict persistent pain development and suggest a role for microRNA in sex-specific pain differences. Mol. Pain 2015, 11, 66. [Google Scholar] [CrossRef] [Green Version]
  60. Linnstaedt, S.D.; Riker, K.D.; Walker, M.G.; Nyland, J.E.; Zimny, E.; Lewandowski, C.; Hendry, P.L.; Damiron, K.; Pearson, C.; Velilla, M.-A.; et al. MicroRNA 320a Predicts Chronic Axial and Widespread Pain Development Following Motor Vehicle Collision in a Stress-Dependent Manner. J. Orthop. Sports Phys. Ther. 2016, 46, 911–919. [Google Scholar] [CrossRef]
  61. Linnstaedt, S.D.; Rueckeis, C.A.; Riker, K.D.; Pan, Y.; Wu, A.; Yu, S.; Wanstrath, B.; Gonzalez, M.; Harmon, E.; Green, P.; et al. MicroRNA-19b predicts widespread pain and posttraumatic stress symptom risk in a sex-dependent manner following trauma exposure. Pain 2019, 161, 47–60. [Google Scholar] [CrossRef]
  62. Elliott, J.M.; Rueckeis, C.A.; Pan, Y.; Parrish, T.B.; Walton, D.M.; Linnstaedt, S.D. microRNA let-7i-5p mediates the relationship between muscle fat infiltration and neck pain disability following motor vehicle collision: A preliminary study. Sci. Rep. 2021, 11, 3140. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, J.; Xue, D.; Wang, X.; Ai, D.; Qin, P. MiR-101 relates to chronic peripheral neuropathic pain through targeting KPNB1 and regulating NF-κB signaling. Kaohsiung J. Med. Sci. 2019, 35, 139–145. [Google Scholar] [CrossRef] [PubMed]
  64. Popiolek-Barczyk, K.; Makuch, W.; Rojewska, E.; Pilat, D.; Mika, J. Inhibition of intracellular signaling pathways NF-κB and MEK1/2 attenuates neuropathic pain development and enhances morphine analgesia. Pharmacol. Rep. 2014, 66, 845–851. [Google Scholar] [CrossRef]
  65. Leinders, M.; Üçeyler, N.; Thomann, A.; Sommer, C. Aberrant microRNA expression in patients with painful peripheral neuropathies. J. Neurol. Sci. 2017, 380, 242–249. [Google Scholar] [CrossRef] [PubMed]
  66. Li, X.; Huang, Y.; Zhang, Y.; He, N. Evaluation of microRNA Expression in Patients with Herpes Zoster. Viruses 2016, 8, 326. [Google Scholar] [CrossRef] [Green Version]
  67. Kalichman, L.; Hunter, D.J. The genetics of intervertebral disc degeneration.Familial predisposition and heritability estimation. Jt. Bone Spine 2008, 75, 383–387. [Google Scholar] [CrossRef]
  68. Cui, S.; Zhou, Z.; Liu, X.; Richards, R.G.; Alini, M.; Peng, S.; Liu, S.; Zou, X.; Li, Z.; Grad, S. Identification and Characterization of Serum microRNAs as Biomarkers for Human Disc Degeneration: An RNA Sequencing Analysis. Diagnostics 2020, 10, 1063. [Google Scholar] [CrossRef]
  69. Dong, W.; Liu, J.; Lv, Y.; Wang, F.; Liu, T.; Sun, S.; Liao, B.; Shu, Z.; Qian, J. miR-640 aggravates intervertebral disc degeneration via NF-κB and WNT signalling pathway. Cell Prolif. 2019, 52, e12664. [Google Scholar] [CrossRef] [Green Version]
  70. Hasvik, E.; Schjølberg, T.; Jacobsen, D.P.; Haugen, A.J.; Grøvle, L.; Schistad, E.I.; Gjerstad, J. Up-regulation of circulating microRNA-17 is associated with lumbar radicular pain following disc herniation. Arthritis Res. Ther. 2019, 21, 186. [Google Scholar] [CrossRef] [Green Version]
  71. van de Merwe, J.P.; Nordling, J.; Bouchelouche, P.; Bouchelouche, K.; Cervigni, M.; Daha, L.K.; Elneil, S.; Fall, M.; Hohlbrugger, G.; Irwin, P.; et al. Diagnostic Criteria, Classification, and Nomenclature for Painful Bladder Syndrome/Interstitial Cystitis: An ESSIC Proposal. Eur. Urol. 2008, 53, 60–67. [Google Scholar] [CrossRef]
  72. Freire, V.S.; Burkhard, F.C.; Kessler, T.M.; Kuhn, A.; Draeger, A.; Monastyrskaya, K. MicroRNAs May Mediate the Down-Regulation of Neurokinin-1 Receptor in Chronic Bladder Pain Syndrome. Am. J. Pathol. 2010, 176, 288–303. [Google Scholar] [CrossRef] [PubMed]
  73. Arai, T.; Fuse, M.; Goto, Y.; Kaga, K.; Kurozumi, A.; Yamada, Y.; Sugawara, S.; Okato, A.; Ichikawa, T.; Yamanishi, T.; et al. Molecular pathogenesis of interstitial cystitis based on microRNA expression signature: miR-320 family-regulated molecular pathways and targets. J. Hum. Genet. 2018, 63, 543–554. [Google Scholar] [CrossRef] [PubMed]
  74. Schaeffer, A.J. Clinical Practice. Chronic Prostatitis and the Chronic Pelvic Pain Syndrome. N. Engl. J. Med. 2006, 355, 1690–1698. [Google Scholar] [CrossRef] [Green Version]
  75. Chen, Y.; Chen, S.; Zhang, J.; Wang, Y.; Jia, Z.; Zhang, X.; Han, X.; Guo, X.; Sun, X.; Shao, C.; et al. Expression profile of microRNAs in expressed prostatic secretion of healthy men and patients with IIIA chronic prostatitis/chronic pelvic pain syndrome. Oncotarget 2018, 9, 12186–12200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liang, Z.; Chen, Y.; Zhao, Y.; Xu, C.; Zhang, A.; Zhang, Q.; Wang, D.; He, J.; Hua, W.; Duan, P. miR-200c suppresses endometriosis by targeting MALAT1 in vitro and in vivo. Stem Cell Res. Ther. 2017, 8, 251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Razi, M.H.; Eftekhar, M.; Ghasemi, N.; Sheikhha, M.H.; Firoozabadi, A.D. Expression levels of circulatory mir-185-5p, vascular endothelial growth factor, and platelet-derived growth factor target genes in endometriosis. Int. J. Reprod. Biomed. 2020, 18, 347–358. [Google Scholar] [CrossRef]
  78. Perrot, S.; Cohen, M.; Barke, A.; Korwisi, B.; Rief, W.; Treede, R.D.; IASP Taskforce for the Classification of Chronic Pain. The IASP classification of chronic pain for ICD-11: Chronic secondary musculoskeletal pain. Pain 2019, 160, 77–82. [Google Scholar] [CrossRef]
  79. Dayer, C.F.; Luthi, F.; Le Carré, J.; Vuistiner, P.; Terrier, P.; Benaim, C.; Giacobino, J.-P.; Léger, B. Differences in the miRNA signatures of chronic musculoskeletal pain patients from neuropathic or nociceptive origins. PLoS ONE 2019, 14, e0219311. [Google Scholar] [CrossRef]
  80. Schaible, H.-G. Mechanisms of Chronic Pain in Osteoarthritis. Curr. Rheumatol. Rep. 2012, 14, 549–556. [Google Scholar] [CrossRef]
  81. Fu, K.; Robbins, S.; McDougall, J. Osteoarthritis: The genesis of pain. Rheumatology 2017, 57, iv43–iv50. [Google Scholar] [CrossRef] [Green Version]
  82. Ashraf, S.; Wibberley, H.; Mapp, P.I.; Hill, R.; Wilson, D.; Walsh, D. Increased vascular penetration and nerve growth in the meniscus: A potential source of pain in osteoarthritis. Ann. Rheum. Dis. 2010, 70, 523–529. [Google Scholar] [CrossRef] [PubMed]
  83. Xie, W.; Su, W.; Xia, H.; Wang, Z.; Su, C.; Su, B. Synovial Fluid MicroRNA-210 as a Potential Biomarker for Early Prediction of Osteoarthritis. BioMed Res. Int. 2019, 2019, 7165406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Skrzypa, M.; Szala, D.; Gablo, N.; Czech, J.; Pajak, J.; Kopańska, M.; Trzeciak, M.; Gargasz, K.; Snela, S.; Zawlik, I. miRNA-146a-5p is upregulated in serum and cartilage samples of patients with osteoarthritis. Pol. J. Surg. 2018, 91, 1–5. [Google Scholar] [CrossRef] [PubMed]
  85. Tian, F.; Wang, J.; Zhang, Z.; Yang, J. LncRNA SNHG7/miR-34a-5p/SYVN1 axis plays a vital role in proliferation, apoptosis and autophagy in osteoarthritis. Biol. Res. 2020, 53, 9. [Google Scholar] [CrossRef] [PubMed]
  86. Brandenburger, T.; Johannsen, L.; Prassek, V.; Kuebart, A.; Raile, J.; Wohlfromm, S.; Köhrer, K.; Huhn, R.; Hollmann, M.W.; Hermanns, H. MiR-34a is differentially expressed in dorsal root ganglia in a rat model of chronic neuropathic pain. Neurosci. Lett. 2019, 708, 134365. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, H.-H.; Zhang, X.-Q.; Wang, W.-Y.; Xue, Q.-S.; Lu, H.; Huang, J.-L.; Gui, T.; Yu, B.-W. Increased Synaptophysin Is Involved in Inflammation-Induced Heat Hyperalgesia Mediated by Cyclin-Dependent Kinase 5 in Rats. PLoS ONE 2012, 7, e46666. [Google Scholar] [CrossRef]
  88. Wang, T.; Hao, Z.; Liu, C.; Yuan, L.; Li, L.; Yin, M.; Li, Q.; Qi, Z.; Wang, Z. LEF1 mediates osteoarthritis progression through circRNF121/miR-665/MYD88 axis via NF-κB signaling pathway. Cell Death Dis. 2020, 11, 598. [Google Scholar] [CrossRef]
  89. Kwak, Y.H.; Kwak, D.-K.; Kim, N.Y.; Kim, Y.J.; Lim, J.S.; Yoo, J.-H. Significant changes in synovial fluid microRNAs after high tibial osteotomy in medial compartmental knee osteoarthritis: Identification of potential prognostic biomarkers. PLoS ONE 2020, 15, e0227596. [Google Scholar] [CrossRef] [Green Version]
  90. Leinders, M.; Üçeyler, N.; Pritchard, R.; Sommer, C.; Sorkin, L. Increased miR-132-3p expression is associated with chronic neuropathic pain. Exp. Neurol. 2016, 283, 276–286. [Google Scholar] [CrossRef] [Green Version]
  91. Huang, Y.; Li, X.; Tao, G.; Zhu, T.; Lin, J. Comparing serum microRNA levels of acute herpes zoster patients with those of postherpetic neuralgia patients. Medicine 2017, 96, e5997. [Google Scholar] [CrossRef]
  92. Moen, A.; Jacobsen, D.; Phuyal, S.; Legfeldt, A.; Haugen, F.; Røe, C.; Gjerstad, J. MicroRNA-223 demonstrated experimentally in exosome-like vesicles is associated with decreased risk of persistent pain after lumbar disc herniation. J. Transl. Med. 2017, 15, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lin, H.; Zhang, W.; Zhou, T.; Li, W.; Chen, Z.; Ji, C.; Zhang, C.; He, F. Mechanism of microRNA-21 regulating IL-6 inflammatory response and cell autophagy in intervertebral disc degeneration. Exp. Ther. Med. 2017, 14, 1441–1444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhao, B.; Yu, Q.; Li, H.; Guo, X.; He, X. Characterization of microRNA expression profiles in patients with intervertebral disc degeneration. Int. J. Mol. Med. 2013, 33, 43–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Guo, W.; Zhang, B.; Sun, C.; Duan, H.; Liu, W.; Mu, K.; Zhao, L.; Li, H.; Dong, Z.; Cui, Q. Circular RNA derived from TIMP2 functions as a competitive endogenous RNA and regulates intervertebral disc degeneration by targeting miR-185-5p and matrix metalloproteinase 2. Int. J. Mol. Med. 2020, 46, 621–632. [Google Scholar] [CrossRef]
  96. Gu, C.-L.; Zhang, Z.; Fan, W.-S.; Li, L.-A.; Ye, M.-X.; Zhang, Q.; Zhang, N.-N.; Li, Z.; Meng, Y.-G. Identification of MicroRNAs as Potential Biomarkers in Ovarian Endometriosis. Reprod. Sci. 2020, 27, 1715–1723. [Google Scholar] [CrossRef]
  97. Burney, R.; Hamilton, A.; Aghajanova, L.; Vo, K.; Nezhat, C.; Lessey, B.; Giudice, L. MicroRNA expression profiling of eutopic secretory endometrium in women with versus without endometriosis. Mol. Hum. Reprod. 2009, 15, 625–631. [Google Scholar] [CrossRef]
  98. Beyer, C.; Zampetaki, A.; Lin, N.-Y.; Kleyer, A.; Perricone, C.; Iagnocco, A.; Distler, A.; Langley, S.R.; Gelse, K.; Sesselmann, S.M.; et al. Signature of circulating microRNAs in osteoarthritis. Ann. Rheum. Dis. 2014, 74, e18. [Google Scholar] [CrossRef] [Green Version]
  99. Cuadra, V.M.B.; González-Huerta, N.C.; Romero-Cordoba, S.L.; Hidalgo-Miranda, A.; Miranda-Duarte, A. Altered Expression of Circulating MicroRNA in Plasma of Patients with Primary Osteoarthritis and In Silico Analysis of Their Pathways. PLoS ONE 2014, 9, e97690. [Google Scholar] [CrossRef]
  100. Okuhara, A.; Nakasa, T.; Shibuya, H.; Niimoto, T.; Adachi, N.; Deie, M.; Ochi, M. Changes in microRNA expression in peripheral mononuclear cells according to the progression of osteoarthritis. Mod. Rheumatol. 2011, 22, 446–457. [Google Scholar] [CrossRef]
  101. Mathé, E.; Nguyen, G.H.; Funamizu, N.; He, P.; Moake, M.; Croce, C.M.; Hussain, S.P. Inflammation regulates microRNA expression in cooperation with p53 and nitric oxide. Int. J. Cancer 2011, 131, 760–765. [Google Scholar] [CrossRef] [Green Version]
  102. Rukov, J.L.; Shomron, N. MicroRNA pharmacogenomics: Post-transcriptional regulation of drug response. Trends Mol. Med. 2011, 17, 412–423. [Google Scholar] [CrossRef] [PubMed]
  103. Pehserl, A.-M.; Ress, A.L.; Stanzer, S.; Resel, M.; Karbiener, M.; Stadelmeyer, E.; Stiegelbauer, V.; Gerger, A.; Mayr, C.; Scheideler, M.; et al. Comprehensive Analysis of miRNome Alterations in Response to Sorafenib Treatment in Colorectal Cancer Cells. Int. J. Mol. Sci. 2016, 17, 2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Finnerty, J.R.; Wang, W.-X.; Hébert, S.; Wilfred, B.R.; Mao, G.; Nelson, P.T. The miR-15/107 Group of MicroRNA Genes: Evolutionary Biology, Cellular Functions, and Roles in Human Diseases. J. Mol. Biol. 2010, 402, 491–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Lu, Q.; Ma, Z.; Ding, Y.; Bedarida, T.; Chen, L.; Xie, Z.; Song, P.; Zou, M.-H. Circulating miR-103a-3p contributes to angiotensin II-induced renal inflammation and fibrosis via a SNRK/NF-κB/p65 regulatory axis. Nat. Commun. 2019, 10, 2145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Sarnico, I.; Branca, C.; Lanzillotta, A.; Porrini, V.; Benarese, M.; Spano, P.F.; Pizzi, M. NF-κB and epigenetic mechanisms as integrative regulators of brain resilience to anoxic stress. Brain Res. 2012, 1476, 203–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Gutierrez, H.; Davies, A.M. Regulation of neural process growth, elaboration and structural plasticity by NF-κB. Trends Neurosci. 2011, 34, 316–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Alivernini, S.; Gremese, E.; McSharry, C.; Tolusso, B.; Ferraccioli, G.; McInnes, I.B.; Kurowska-Stolarska, M. MicroRNA-155—At the Critical Interface of Innate and Adaptive Immunity in Arthritis. Front. Immunol. 2018, 8, 1932. [Google Scholar] [CrossRef] [Green Version]
  109. Hu, J.; Huang, S.; Liu, X.; Zhang, Y.; Wei, S.; Hu, X. miR-155: An Important Role in Inflammation Response. J. Immunol. Res. 2022, 2022, 7437281. [Google Scholar] [CrossRef]
  110. Chamorro-Jorganes, A.; Araldi, E.; Suárez, Y. MicroRNAs as pharmacological targets in endothelial cell function and dysfunction. Pharmacol. Res. 2013, 75, 15–27. [Google Scholar] [CrossRef] [Green Version]
  111. Zingale, V.D.; Gugliandolo, A.; Mazzon, E. MiR-155: An Important Regulator of Neuroinflammation. Int. J. Mol. Sci. 2021, 23, 90. [Google Scholar] [CrossRef]
  112. Ou, G.-Y.; Lin, W.-W.; Zhao, W.-J. Neuregulins in Neurodegenerative Diseases. Front. Aging Neurosci. 2021, 13, 662474. [Google Scholar] [CrossRef] [PubMed]
  113. Gaudet, A.D.; Fonken, L.K.; Ayala, M.T.; Maier, S.F.; Watkins, L.R. Aging and miR-155 in mice influence survival and neuropathic pain after spinal cord injury. Brain Behav. Immun. 2021, 97, 365–370. [Google Scholar] [CrossRef] [PubMed]
  114. Duan, Z.; Zhang, J.; Li, J.; Pang, X.; Wang, H. Inhibition of microRNA-155 Reduces Neuropathic Pain During Chemotherapeutic Bortezomib via Engagement of Neuroinflammation. Front. Oncol. 2020, 10, 416. [Google Scholar] [CrossRef]
  115. Tan, Y.; Yang, J.; Xiang, K.; Tan, Q.; Guo, Q. Suppression of MicroRNA-155 Attenuates Neuropathic Pain by Regulating SOCS1 Signalling Pathway. Neurochem. Res. 2014, 40, 550–560. [Google Scholar] [CrossRef]
  116. Roush, S.; Slack, F.J. The let-7 family of microRNAs. Trends Cell Biol. 2008, 18, 505–516. [Google Scholar] [CrossRef]
  117. Esquela-Kerscher, A.; Slack, F. Oncomirs—microRNAs with a role in cancer. Nat. Cancer 2006, 6, 259–269. [Google Scholar] [CrossRef]
  118. Cho, K.J.; Song, J.; Oh, Y.; Lee, J.E. MicroRNA-Let-7a regulates the function of microglia in inflammation. Mol. Cell. Neurosci. 2015, 68, 167–176. [Google Scholar] [CrossRef]
  119. He, Y.; Wang, Z.J. Let-7 microRNAs and Opioid Tolerance. Front. Genet. 2012, 3, 110. [Google Scholar] [CrossRef] [Green Version]
  120. Yun, Z.; Wang, Y.; Feng, W.; Zang, J.; Zhang, D.; Gao, Y. Overexpression of microRNA-185 alleviates intervertebral disc degeneration through inactivation of the Wnt/β-catenin signaling pathway and downregulation of Galectin-3. Mol. Pain 2020, 16, 1744806920902559. [Google Scholar] [CrossRef]
  121. Huang, A.; Ji, L.; Huang, Y.; Yu, Q.; Li, Y. miR-185-5p alleviates CCI-induced neuropathic pain by repressing NLRP3 inflammasome through dual targeting MyD88 and CXCR4. Int. Immunopharmacol. 2022, 104, 108508. [Google Scholar] [CrossRef]
  122. Cheng, Z.; Qiu, S.; Jiang, L.; Zhang, A.; Bao, W.; Liu, P.; Liu, J. MiR-320a is Downregulated in Patients with Myasthenia Gravis and Modulates Inflammatory Cytokines Production by Targeting Mitogen-activated Protein Kinase 1. J. Clin. Immunol. 2012, 33, 567–576. [Google Scholar] [CrossRef]
  123. Wu, J.; Sabirzhanov, B.; Stoica, B.A.; Lipinski, M.M.; Zhao, Z.; Zhao, S.; Ward, N.; Yang, D.; Faden, A.I. Ablation of the transcription factors E2F1-2 limits neuroinflammation and associated neurological deficits after contusive spinal cord injury. Cell Cycle 2015, 14, 3698–3712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Kopańska, M.; Szala, D.; Czech, J.; Gabło, N.; Gargasz, K.; Trzeciak, M.; Zawlik, I.; Snela, S. MiRNA expression in the cartilage of patients with osteoarthritis. J. Orthop. Surg. Res. 2017, 12, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Li, X.; Gibson, G.; Kim, J.-S.; Kroin, J.; Xu, S.; van Wijnen, A.J.; Im, H.-J. MicroRNA-146a is linked to pain-related pathophysiology of osteoarthritis. Gene 2011, 480, 34–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Nakasa, T.; Miyaki, S.; Okubo, A.; Hashimoto, M.; Nishida, K.; Ochi, M.; Asahara, H. Expression of microRNA-146 in rheumatoid arthritis synovial tissue. Arthritis Care Res. 2008, 58, 1284–1292. [Google Scholar] [CrossRef] [Green Version]
  127. Ji, R.-R.; Chamessian, A.; Zhang, Y.-Q. Pain regulation by non-neuronal cells and inflammation. Science 2016, 354, 572–577. [Google Scholar] [CrossRef] [Green Version]
  128. Cho, C.; Michailidis, V.; Martin, L.J. Revealing brain mechanisms of mTOR-mediated translational regulation: Implications for chronic pain. Neurobiol. Pain 2018, 4, 27–34. [Google Scholar] [CrossRef]
  129. Totsch, S.K.; Sorge, R.E. Immune system involvement in specific pain conditions. Mol. Pain 2017, 13. [Google Scholar] [CrossRef] [Green Version]
  130. Malcangio, M. Role of the immune system in neuropathic pain. Scand. J. Pain 2019, 20, 33–37. [Google Scholar] [CrossRef]
  131. Khan, N.; Smith, M.T. Neurotrophins and Neuropathic Pain: Role in Pathobiology. Molecules 2015, 20, 10657–10688. [Google Scholar] [CrossRef]
  132. Wood, P.B. Role of central dopamine in pain and analgesia. Expert Rev. Neurother. 2008, 8, 781–797. [Google Scholar] [CrossRef] [PubMed]
  133. Elman, I.; Borsook, D. Common Brain Mechanisms of Chronic Pain and Addiction. Neuron 2016, 89, 11–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. LeBlanc, D.M.; McGinn, M.A.; Itoga, C.; Edwards, S. The affective dimension of pain as a risk factor for drug and alcohol addiction. Alcohol 2015, 49, 803–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Liu, K.; Tong, H.; Li, T.; Wang, X.; Chen, Y. Research progress in molecular biology related quantitative methods of MicroRNA. Am. J. Transl. Res. 2020, 12, 3198–3211. [Google Scholar]
  136. Tam, S.; De Borja, R.; Tsao, M.; McPherson, J. Robust global microRNA expression profiling using next-generation sequencing technologies. Lab. Investig. 2014, 94, 350–358. [Google Scholar] [CrossRef] [Green Version]
  137. Dweep, H.; Gretz, N.; Sticht, C. miRWalk Database for miRNA–Target Interactions. Methods Mol. Biol. 2014, 1182, 289–305. [Google Scholar] [CrossRef]
  138. Heberle, H.; Meirelles, G.V.; Da Silva, F.R.; Telles, G.P.; Minghim, R. InteractiVenn: A Web-Based Tool for the Analysis of Sets through Venn Diagrams. BMC Bioinform. 2015, 16, 169. [Google Scholar] [CrossRef]
  139. Su, G.; Morris, J.H.; Demchak, B.; Bader, G.D. Biological network exploration with Cytoscape 3. Curr. Protoc. Bioinform. 2014, 47, 8.13.1–8.13.24. [Google Scholar] [CrossRef] [Green Version]
  140. Stöckel, D.; Kehl, T.; Trampert, P.; Schneider, L.; Backes, C.; Ludwig, N.; Gerasch, A.; Kaufmann, M.; Gessler, M.; Graf, N.; et al. Multi-omics enrichment analysis using the GeneTrail2 web service. Bioinformatics 2016, 32, 1502–1508. [Google Scholar] [CrossRef]
  141. R Development Core Team. R: A Language and Environment for Statistical Computing. Available online: https://www.R-project.org/ (accessed on 19 November 2020).
Figure 1. The miRNA–mRNA regulatory network. Red circles represent miRNAs and blue circles represent target genes.
Figure 1. The miRNA–mRNA regulatory network. Red circles represent miRNAs and blue circles represent target genes.
Ijms 23 06016 g001
Figure 2. KEGG pathways related to cellular processes significantly associated with miRNA targets in CPP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Figure 2. KEGG pathways related to cellular processes significantly associated with miRNA targets in CPP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Ijms 23 06016 g002
Figure 3. KEGG pathways related to cellular processes significantly associated with miRNA targets in CSP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Figure 3. KEGG pathways related to cellular processes significantly associated with miRNA targets in CSP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Ijms 23 06016 g003
Figure 4. KEGG pathways related to diseases significantly associated with miRNA targets in CPP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Figure 4. KEGG pathways related to diseases significantly associated with miRNA targets in CPP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Ijms 23 06016 g004
Figure 5. KEGG pathways related to diseases significantly associated with miRNA targets in CSP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Figure 5. KEGG pathways related to diseases significantly associated with miRNA targets in CSP (within each group, pathways are arranged according to their p-values, in descending order). Adjusted p-values were calculated using Benjamini-Yekutieli method.
Ijms 23 06016 g005
Table 1. The IASP classification of chronic pain (modified by [3]).
Table 1. The IASP classification of chronic pain (modified by [3]).
Chronic PainChronic Primary PainChronic widespread pain
Complex regional pain syndrome
Chronic primary headache or orofacial pain
Chronic primary visceral pain
Chronic primary musculoskeletal pain
Chronic Secondary PainChronic cancer-related pain
Chronic postsurgical or posttraumatic pain
Chronic neuropathic pain
Chronic secondary headache or orofacial pain
Chronic secondary visceral pain
Chronic secondary musculoskeletal pain
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sabina, S.; Panico, A.; Mincarone, P.; Leo, C.G.; Garbarino, S.; Grassi, T.; Bagordo, F.; De Donno, A.; Scoditti, E.; Tumolo, M.R. Expression and Biological Functions of miRNAs in Chronic Pain: A Review on Human Studies. Int. J. Mol. Sci. 2022, 23, 6016. https://doi.org/10.3390/ijms23116016

AMA Style

Sabina S, Panico A, Mincarone P, Leo CG, Garbarino S, Grassi T, Bagordo F, De Donno A, Scoditti E, Tumolo MR. Expression and Biological Functions of miRNAs in Chronic Pain: A Review on Human Studies. International Journal of Molecular Sciences. 2022; 23(11):6016. https://doi.org/10.3390/ijms23116016

Chicago/Turabian Style

Sabina, Saverio, Alessandra Panico, Pierpaolo Mincarone, Carlo Giacomo Leo, Sergio Garbarino, Tiziana Grassi, Francesco Bagordo, Antonella De Donno, Egeria Scoditti, and Maria Rosaria Tumolo. 2022. "Expression and Biological Functions of miRNAs in Chronic Pain: A Review on Human Studies" International Journal of Molecular Sciences 23, no. 11: 6016. https://doi.org/10.3390/ijms23116016

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop