Next Article in Journal
Dysregulation of Lipid Metabolism in Aging Meibomian Glands and Its Molecular Markers
Previous Article in Journal
Macrophage-Myofibroblast Transition Contributes to Myofibroblast Formation in Proliferative Vitreoretinal Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Common miRNAs of Osteoporosis and Fibromyalgia: A Review

1
Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France
2
Inserm 1107, Neuro-Dol, University Clermont Auvergne, F-63000 Clermont-Ferrand, France
3
Rheumatology Department, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(17), 13513; https://doi.org/10.3390/ijms241713513
Submission received: 10 July 2023 / Revised: 28 August 2023 / Accepted: 29 August 2023 / Published: 31 August 2023

Abstract

:
A significant clinical association between osteoporosis (OP) and fibromyalgia (FM) has been shown in the literature. Given the need for specific biomarkers to improve OP and FM management, common miRNAs might provide promising tracks for future prevention and treatment. The aim of this review is to identify miRNAs described in OP and FM, and dysregulated in the same direction in both pathologies. The PubMed database was searched until June 2023, with a clear mention of OP, FM, and miRNA expression. Clinical trials, case–control, and cross-sectional studies were included. Gray literature was not searched. Out of the 184 miRNAs found in our research, 23 are shared by OP and FM: 7 common miRNAs are dysregulated in the same direction for both pathologies (3 up-, 4 downregulated). The majority of these common miRNAs are involved in the Wnt pathway and the cholinergic system and a possible link has been highlighted. Further studies are needed to explore this relationship. Moreover, the harmonization of technical methods is necessary to confirm miRNAs shared between OP and FM.

1. Introduction

Osteoporosis (OP) is a systemic disease that affects skeletal architecture. It is characterized by decreased bone mineral density (BMD) and increased risk of fragility fractures, disability, and impaired quality of life. OP prevalence increases with age at a rate of 19.1–23.5% in women after 50 years, and 5.9–7.2% in men, in whom it still remains underdiagnosed [1,2], and affects millions of people worldwide. OP is a complex multifactorial pathology that may remain undetected for a long time until a fracture occurs. It is diagnosed clinically and via dual-energy X-ray absorptiometry (DEXA). Mechanical, metabolic, and hormonal influences, aging, menopause, genetic predisposition, and environment, including nutrition, are the main factors for OP development. Although cost-effective therapeutic interventions to reduce fractures have been developed, there are a number of gaps in the general management of OP, and preventive measures need to be extended [3,4].
The fibromyalgia syndrome (FM) consists of chronic symptoms of moderate to severe intensity with chronic widespread (nociplastic) pain, associated with fatigue, cognitive and sleep disorders, and numerous somatic complaints [5,6,7,8]. Its manifestations are heterogeneous at the clinical, physical, social, and psychological levels, and treatment failures are frequent [9,10]. The prevalence of FM in the general population is estimated as 1.78% (95% confidence interval: 1.65–1.92) [11] with a female predominance, and, like in OP, it remains underestimated in men. The detection of FM follows the American College of Rheumatology recommendations [12], and, for its management, international guidelines recommend non-pharmacological approaches (exercise) in the first instance, and then drug treatment for comorbidities and pain, but quality of life is often impaired. Although a number of predictive factors of FM including biomarkers have been suggested [5], no specific biomarker has so far been identified.
Clinically, associations between FM and OP have been shown in the literature. Since the earlier works published in the 90s [13,14], FM has been suggested in a number of studies to be associated with an increased risk of OP [1,15,16]. In a meta-analysis, BMD at the lumbar spine was decreased in FM compared with normal individuals, stressing that the risk assessment of OP should be systematically performed [15]. FM shares common risk factors with OP, including age, gender, hygiene, dietary habits/lifestyle factors, a low level of physical activity, and hormonal factors [16]. A meta-analysis [17] demonstrated that BMD was significantly lower in FM patients, in Caucasians, and in female populations. A population-based case–control electronic study recently showed a significant association (coefficient correlation 0.55; p < 0.001) between OP and FM with a large database [1]. The authors underlined the need for the detection of predisposing factors for OP in FM patients, and advised the implementation of prevention measures (dietary supplements, resistance or weight-bearing exercise, anti-OP drugs). These, in order to maintain a satisfactory quality of life, reduce both the occurrence rate and severity of OP and its complications, such as fractures.
OP and FM have each been described as being associated with a number of possible biomarkers, including epigenetic markers and microRNAs (miRNAs) [18,19], known to play important roles in regulating gene expression. miRNAs have been a focus of research over the last years in OP and in FM. miRNAs, a class of non-coding RNAs 18 to 25 nucleotides long, are known to control gene expression at the post-transcriptional level [20] by forming an RNA-induced silencing complex which directly modulates the gene expression of mRNA genes [21]. miRNAs could regulate more than 60% of protein-coding genes and are therefore involved in most biological processes [22]. Epigenetics play important roles in bone metabolism and bone remodeling. An abnormal regulation may induce OP development and a number of miRNAs have been identified [23,24,25,26]. Likewise, miRNAs have been studied and described in FM [18,27]. Considering the need for specific biomarkers to improve OP and FM management, common miRNAs might provide promising tracks for future prevention and treatment. In order to find significant circulating predictive markers and potential new therapeutic targets for FM and OP, the objective of this review is to identify common miRNAs with similar regulation in OP and FM. To the best of our knowledge, there is currently no previous review on the common epigenetic markers of OP and FM.

2. Results and Discussion

2.1. Included Studies

Our search (Figure 1) retrieved 4922 potentially relevant records for the current scoping review, 232 for FM and 4690 for OP. After removing duplicates, the titles and abstracts of the remaining 1144 (36 for FM and 1108 for OP) were screened by two reviewers (SP, MD). After discarding records not conforming with the inclusion criteria, the full texts of the eligible papers were reviewed and 137 studies were included in this scoping review, 8 for FM [28,29,30,31,32,33,34,35] and 129 for OP [23,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163].
In these 137 publications, there was a total of 189 dysregulated miRNAs (versus healthy volunteers (HV)): 46 miRNAs in FM and 166 in OP (Figure 2 and Figure 3, Tables S1 and S2).
In 45/137 articles, 23 miRNAs were common for FM and OP (Figure 2, Table 1): 14 articles showing 7 common miRNAs dysregulated in the same direction, and 40 articles showing 16 miRNAs dysregulated in the opposite direction (n = 9) or discordant (n = 7 dysregulated in the same or in the opposite direction in FM and OP).
In 14/45 articles, according to our objective to identify common miRNAs with a similar regulation, seven miRNAs were dysregulated (up- or downregulation) in the same direction for both pathologies: three miRNAs were upregulated (hsa-miR-9-(3p or 5p), hsa-miR-128-(3p), and hsa-335- 5p), and four were downregulated (hsa-miR-1-(3p), hsa-let-7a-(3p or 5p), hsa-miR-29a-3p, and hsa-miR-328-3p).

2.2. Characteristics of Studies with Common miRNAs in FM and OP, and With Regulation in the Same Direction

The 45 studies were published between 2012 and 2022, mainly with Caucasian participants for FM and Asian participants for OP (Tables S3 and S4).

2.2.1. Sample Size

In the 14 articles related to the seven common miRNAs regulated in the same direction (up or down), three of the five FM studies had less than 50 participants, the largest (n = 74) with 49 FM patients and 25 HV [30], and the smallest (n = 18) with 10 FM and 8 HV [31]. For the nine articles for OP, two studies included more than 100 persons, four between 50 and 80 participants, and three less than 50. The largest study (n = 116) included 76 low-BMD participants with or without fractures and treatment, and 40 HV [43], and the smallest (n = 6) included 3 OP and 3 HV [37].
In the 45 articles, the majority of FM studies (71%: n = 5) had less than 50 participants, the largest (n = 74) with 49 FM patients and 25 HV [30], and the smallest (n = 18) with 10 FM and 8 HV [31]. For OP, six studies included more than 100 persons, six between 50 and 80 participants, and twenty-five less than 50. One [55] did not mention the sample size. The largest study (n = 161) included 82 OP and 79 HV [65], and the smallest (n = 6) 3 OP and 3 HV [37,47].

2.2.2. Age and Gender

Concerning the seven miRNAs regulated in the same direction (up or down), all cohorts in FM patients were around 50 years old, except one [28] that did not mention the age. In OP, the patients were over 50 years old, with 70-year-old persons in five studies [23,36,37,41,43] and no mention of age in several [38,39,40]. In FM, all studies but one [32]—with only 30% men—included only females. In OP, six out of nine studies for OP were 100% women. One study included both genders, with 60% women [42]. The remaining two studies did not mention gender [39,41].
In the 45 articles, the FM patients were around 50 years old, and OP patients were over 50 years old, with 80-year-old persons in 2 studies [56,59]. Age was not always mentioned in FM [28] or in OP [38,39,40,46,48,55,70,72]. In FM, all studies but one [32]—with only 30% men—included only females. In OP, 25 of 38 studies for OP had only women. For studies that included both genders, the majority was with 60 to 90% women. One study included the same number of men and women for patient and HV groups [56]. The remaining nine studies did not mention gender.

2.2.3. Tissue Sample, Extraction, and Detection Method

For the seven miRNAs regulated in the same direction (up or down), concerning extraction, four out of five FM studies were on circulating miRNAs: serum [28,29,32], cerebrospinal fluid [31], or whole blood [30]. Three of the studies used extraction kits from Qiagen, one from Thermo Fisher, and one did not specify the kit used. Concerning detection, qRT-PCR was used, but, in one study [32], a multiplex assay was used. Concerning extraction in OP, 29% of OP studies (n = 4) used circulating miRNAs with serum (n = 4). Other studies used whole blood (n = 3), or other tissues specific to bone, like human-bone-marrow-derived mesenchymal stem cells (hBMSCs) (n = 1) and bone (n = 1). In total, 67% (n = 6) of the studies used extraction kits from Thermo Fisher, the other articles used Qiagen. Three of the four articles with sequencing are found in the nine OP articles where the regulation is in the same direction. A total of 67% (n = 6) used qRT-PCR as a detection method.
In the 45 articles, the extraction was realized with Qiagen or Thermo Fisher (Invitrogen, Ambion) kits, and the detection method was qRT-PCR. Concerning extraction in FM, 57% of FM studies (n = 4) were on circulating miRNAs in the serum [28,29,32], cerebrospinal fluid [31], white blood cells [33], peripheral blood monocellular cells [34], and whole blood [30]. Overall, 57% (n = 4) of the studies used extraction kits from Qiagen, two studies used Thermo Fisher, and one did not specify the kit used. Concerning detection, qRT-PCR was used, but one study [32] used a multiplex assay.
Concerning extraction, in OP, 55% of OP studies (n = 21) used circulating miRNAs, with serum (n = 15) and plasma (n = 6), or whole blood (n = 5) and circulating monocytes (n = 1). Other tissues specific to bone were used like human-bone-marrow-derived mesenchymal stem cells (hBMSCs) (n = 6) and bone (n = 3); two studies [56,59] used both bone and serum. In total, 53% (n = 20) of the studies used extraction kits from Thermo Fisher, 42% (n = 16) used Qiagen, one study used both [37], and one did not specify the kit used [61]. Overall, 86% (n = 33) of OP studies used qRT-PCR as the detection method, only four studies performed sequencing, and one study performed next-generation sequencing (NGS) [53].

2.3. Discussion

Low BMD and OP have been shown to be linked to FM [1,13,14,15,16,17]. In order to better characterize the association between both pathologies, the aim of this review was to identify if there are miRNAs common to FM and OP. It also aimed to specify which miRNAs are regulated in the same direction, and to suggest common biomarkers.
A total of 23 common miRNAs were retrieved in the literature. Fifteen of these are described to target and modulate the Wingless integration site (Wnt) pathway (miR-1-(3p), let-7a-(3p or 5p), miR-9-(3p or 5p), miR-21-5p, miR-29a-3p, miR-107, miR-133a, miR-139-5p, miR-145-(3p or 5p), miR-148a-(3p), miR-186-5p, miR-320a, miR-328-3p, miR-335-5p, and miR-338-3p) [164,165,166,167]. Seven miRNAs modulate the cholinergic system (miR-7-5p, miR-9-(3p or 5p), miR-128-(3p), miR-148a-(3p), miR-186-5p, miR-328-3p, and miR-532-(3p or 5p)), known as “CholinomiR” [30,168]. It is interesting to note that three miRNAs are upregulated and four downregulated in FM and in OP, and they target the Wnt pathway in bone studies, and the cholinergic system in FM studies.
The Wnt system is composed of Wnt proteins that are involved in many cellular processes, ranging from cell-fate determination to stem-cell renewal, and dysregulated Wnt signaling is involved in many human pathologies [169]. Wnt pathways display numerous cross connections that negatively or positively regulate each other, forming a mutual regulatory network.
At the bone level, the Wnt pathway is the most important regulatory pathway; it has a direct effect on skeletal remodeling, regulating bone mass via bone forming osteoblasts, old bone reabsorbing osteoclasts, and progenitor cells responsible for the maintenance of bone-marrow-derived mesenchymal stem cells (BMSCs) [170]. Multiple genes are also targeted, and involved in the regulation of bone, including cyclin D1, RUNX2, bone sialoprotein, sclerostin, Dickkopf 1 and 2, secreted FZ-related protein, osteoprotegerin, osterix, myocyte enhancer factor 2C, osteocalcin, or osteopontin [165,170]. Wnt signaling plays a central regulatory role during embryonic development and in the adult osteogenic differentiation of mesenchymal stem cells. Alterations of this system are accompanied by impaired bone healing, autoimmune disease, osteoporosis, and malignant degeneration. Wnt factors have been suggested as potential future therapeutics to help bone healing after trauma in endocrine or orthopedic situations [171].
The Wnt pathway is ubiquitous and involved in other domains like muscle function [172] or chronic fatigue syndrome (CFS) [173]. FM and CSF are flip sides of almost identical chronic conditions. Patients with CFS have many similar symptoms to those with fibromyalgia—brain fog, fatigue, headache, and poor sleep. Dysregulated Wnt/β-catenin signaling has been shown to cause oxidative stress in animals with CFS, and produce reactive oxygen species (ROS) and aberrations in cross-talks between Wnt, Redox, and NF-kB pathways. The Wnt pathway is also involved in pain sensitization, and neuropathic and bone-cancer-induced pain [174]. Pain accompanies OP especially after trauma and fracture; ensuing central sensitization may lead, in some patients, to neuropathic pain (with burning, stabbing, itching, allodynia, and hyperalgesia). While the origin of FM remains unclear, the central sensitization of pain [175] and the diffuse nociplastic musculoskeletal type of pain are landmarks of FM [6]. In a study centered on an experimental model of FM [174], it has been shown that the Wnt/β-catenin pathway is involved in the release of brain-derived neurotrophic factor from the spinal microglia. This observation suggests that the modulation of this pathway plays a key role in the activation of the nociceptive pathway in the spinal cord [174]. In the same way, the activated Wnt signaling pathway in neuropathic pain [176] modulates the expression of the glutamate receptor, resulting in synaptic plasticity and central sensitization [176].
The cholinergic system is the other system that is expressed by the common miRNAs in FM and OP. Cholinergic mechanisms may play an important role in the pathophysiology and severity of FM [30], especially via the vagus nerve. The vagus nerve is a major cholinergic component of the parasympathetic system, a mixed nerve containing 80% afferent and 20% efferent fibers, which controls the neuro-digestive, vascular, and immune systems. Non-invasive vagus nerve stimulation is even considered today as a potential adjunct treatment for FM [177], since FM involves a dysregulation of the autonomic (high sympathetic tone) and immune (enhanced pro-inflammatory activity and cytokines) systems. Another link between FM and the cholinergic system is medication: a cross-sectional study [178] highlighted that one of the most frequently used and effective FM treatments is amitriptyline, a strong anticholinergic molecule [178].
The cholinergic system is also involved in the health status of bone [179], and cholinergic components (with adrenergic ones, the other branch of the autonomic nervous system) play an important role in bone remodeling. Bone loss associated with OP could be due to local alterations/inhibitions in cholinergic activity, but this has been scarcely studied so far. Clinical studies have shown that bone is also associated with the function of cholinergic-regulated tissues like the hypothalamus [180] and those outside the nervous system in non-neuronal cells.
Common miRNAs have specific signatures in FM and OP (Figure 4, Table 2).
miR-1-(3p), involved in myoblast differentiation, has been suggested to be downregulated in FM because of decreased physical activity in this pathology. miR-1(3p) also modulates Brain Derived Neurotrophic Factor (BDNF) expression in skeletal muscle where it inhibits myogenic differentiation [28,37], and in OP [36]. miR-1-(3p) has a specific target with Secreted Frizzled Related Protein 1 (SFRP1), part of the Wnt signaling system, and balances the osteogenesis and adipogenesis of mesenchymal stem cells (MSCs). Overall, downregulated miR-1-(3p) leads to decreased bone formation and diminished myogenesis [28,36,37].
let-7a-(3p or 5p), downregulated in both diseases [29,38,41,73], plays a role in nerve fiber pathology [35] and regulates pain pathways via the endogenous opioid system in FM [29]. It has a close association with the Wnt pathway in OP [37,41].
miR-9 (3p or 5p) is upregulated in both pathologies. As a cholinomiRNA, it may shift inflammatory processes (Janus kinase 2 expression)—possibly linked to pain—via the modulation of the systemic cholinergic system in FM [30]. It binds to the Wnt pathway, and has a deleterious effect on bone quality [38], skeletal cell proliferation, and differentiation [38,39].
miR-29a-3p is downregulated and is associated with the diminution of β-catenin and with the inactivation of canonical Wnt signaling, leading to OP [23]. In FM, miR-29a-3p expression is reduced compared to healthy controls [31].
miR-128-3p, which is upregulated, is an inhibitor of bone formation via sirtuine 6 (SIRT6) expression [40], and is a cholinomiRNA [30] that modulates the systemic cholinergic system in FM. miR-328-3p is also a cholinomiRNA, which is downregulated, and is associated with osteoblast differentiation in OP [41].
mir-335-5p, which is upregulated, is the only miRNA that was significantly expressed in 105 FM patients compared to 54 controls [32]. mir-335-5p activates Wnt signaling and promotes osteogenic differentiation by downregulating Dickkopf 1 (DKK1), a soluble inhibitor of the Wnt signaling pathway. Two studies with 26 and 39 OP patients compared to control subjects showed miR-335-5p as being upregulated in OP with vertebral/fragility fractures [42,43].
With these seven common miRNAs up- or downregulating in the same direction in FM and OP, the possibility of a link between the Wnt pathway and the cholinergic system could open new avenues for research on the prevention and management of both pathologies. Preclinical studies have highlighted a cholinergic induction of Wnt during infection or immune activation [181,182]. The coordinated activities between acetylcholine receptors and Wnt signaling seem to be conserved in evolutionary terms, and are found in mice [183]. There seems to be a cholinergic–Wnt signaling axis, which can intervene in homeostasis regulation [181], but has not been looked for in OP or FM.
Although we highlighted a number of miRNAs common in FM and OP, there are a number of limitations in this review. The regulation of miRNAs may often be contradictory, even in the same pathology. These mismatches can be explained by differences between studies. From one study to another, the number of patients in the cohort but also in the tissue samples, and technical issues with extraction and detection techniques all vary. miRNAs in FM appear to have less contradictions than in OP, but less research has been carried out on this topic. We noticed that the more studies report miRNA expression, the more contradictory results are obtained.
Another limitation in the interpretation of our results is the ethnical origin of the patients. Indeed, the majority of articles included Asian patients, and results cannot be extrapolated as representing Caucasian characteristics; in addition, there are no studies in African patients or other ethnical groups. While OP is largely reported in women, especially after menopause, OP remains underestimated in men where it usually presents at a later age. Likewise, mostly women seek advice for FM symptoms, while men are less present in pain clinics for this pathology and are poorly represented in publications on FM. This present review underlines the paucity of miRNA studies that include men; such a gap needs to be addressed, since gender-specific differences in miRNAs expression (in the quantity of expression and type of miRNAs) have been described in several pathologies, and such a difference in OP and FM could suggest the development of sex-specific therapeutic strategies.
There is also a need to have prospective longitudinal rather than transversal studies in order to follow FM patients over a number of years, and to evaluate, thoroughly, their bone health and the early presence of OP. Future clinical research should include larger cohorts of patients with a wider range of ethnic representation, more men, and a systematic report on comorbidities that could influence the given pathological condition. Bioinformatics and artificial intelligence can be useful tools in this context to validate miRNAs and identify the predictive value of these common biomarkers for the better diagnosis and management of both pathologies. These miRNAs play a pivotal role in the pathogenesis of FM and OP and are associated with cardinal symptoms, making them interesting potential therapeutic molecules to target.
Finally, some miRNAs are not detected in both pathologies, but it is not certain whether they are present or not, or shared between both diseases, as miRNAs are often prescreened and the choice is based on what researchers want to analyze [184]. Some miRNAs are not secreted by cells, and therefore cannot be detected in circulating miRNAs studies. While qRT-PCR is a sensitive and specific reference method that allows the detection of individual miRNAs or a panel of miRNAs, new detection methods now exist, including deep sequencing. These may help in the future to detect a much wider range and identify new miRNAs, but such techniques are expensive and their development is self-limiting.

3. Materials and Methods

3.1. Review Protocol

This is a systematic/scoping review that was conducted and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) Guidelines [185].

3.2. Eligibility Criteria

Peer-reviewed journal articles were included if they involved the comparison of miRNA expression between HV and patients with OP or FM. An investigation of the miRNAs profiles was followed as a primary or secondary outcome, with any type of biological sample. Clinical trials, case–control, and cross-sectional studies were included. Reviews, meta-analyses, comments, method validations, meeting abstracts, in vitro studies, and animal studies were excluded. All articles that did not clearly mention FM, OP, or miRNA expression were excluded. Database-based studies were not included. There was no age limit nor a minimum number for the population, no specific requirement regarding the year of publication, and studies had to be available in English.

3.3. Information Sources and Search Strategy

In order to identify potentially relevant articles, the online database PubMed was searched until June 2023 with the following keywords: (“fibromyalgia” OR “fibromyalgia syndrome” OR “osteoporosis” OR “post-menopausal osteoporosis”) AND (“miRNA” OR “microRNA” OR “sequencing microRNA” OR “micro RNA” OR “mirRNAs OR “mirs” OR “mir”). The gray literature was not searched nor included. The medical subject heading (MeSH) was used to increase the sensitivity of the systematic search. The reference list of all the full-text articles selected after the screening and the list of articles citing these articles were hand-searched for titles not identified with the previous methods.

3.4. Study Selection Process

Abstracts were obtained for all the studies identified during the electronic and hand-searches, after having removed duplicates. Two reviewers (SP, MD) screened titles and abstracts in the first phase, and full-text copies in the second phase independently to eliminate articles that clearly failed to meet the eligibility criteria. Full-text copies were obtained for all the selected studies.

3.5. Data Charting Process and Synthesis

Predetermined data (including first author, publication year, number for the population, ethnicity, mean age, gender, menopause, age of the disease, comorbidities, biological sample, extraction kit, detection method, study design...) were extracted from each study by two reviewers (SP and MD) and arranged into data tables. The miRNAs cited and explored in the included studies were listed in tables specifying their expression (upregulating or downregulating), according to the targeted pathology (FM or OP). No quality assessment of the included articles took place, which was in accordance with the available guidelines on scoping reviews [185]. We used RNAcentral (http://rnacentral.org), miRBase (http://www.mirbase.org), and Rfam (http://rfam.org) databases (accessed on 18 May 2023) to identify signaling pathways and systems potentially altered by the miRNAs differentially expressed in both pathologies (FM and OP).
According to the nomenclature of the miRNAs, we paid attention to miRNAs originating from the same precursor, the most abundant being named “5p” and the least abundant “3p”. Some articles did not, however, specify the strand. If the literature reported one or the other strand, we indicated it (3p or 5p).

4. Conclusions

Collective data of this review show that a number of common miRNAs in FM and OP have been identified. These are involved in the Wnt pathway for OP and in the cholinergic system for FM. A substantial link is still missing to evaluate the real miRNA impact on Wnt dysregulation in FM and on cholinergic system alterations in OP. Research on this possible link is important since it has been described in animal species with intestinal dysfunction and could be investigated in musculoskeletal conditions. Further research is also warranted on harmonizing techniques or on the choice of tissue for miRNA analysis. Our review underlined a number of gaps linked to the large heterogeneity of methods. We recommend further studies in order to strengthen the epigenetic knowledge of the FM and OP association, and how they are interwoven, to prevent and better manage these pathologies using common predictive biomarkers. Clinical data have reported the increased risk of developing OP in FM patients. The identification of common miRNAs would provide predictive factors for limiting the double burden of FM and OP in aging.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms241713513/s1.

Author Contributions

Conceptualization, N.M. and M.-E.P.; validation, S.P., M.D., N.M., V.M. and M.-E.P.; formal analysis, S.P. and M.D.; investigation, S.P., M.D. and M.-E.P.; resources N.M.; data curation, S.P. and M.D.; writing—original draft preparation, S.P.; writing—review and editing, S.P., M.D., N.M., V.M. and M.-E.P.; visualization, S.P.; supervision, M.-E.P.; project administration, N.M. 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

The original data of this present study are available from the corresponding authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lichtenstein, A.; Tiosano, S.; Comaneshter, D.D.; Cohen, A.; Amital, H. A Case-Control Analysis of the Associations between Fibromyalgia Syndrome and Osteoporosis. Isr. Med. Assoc. J. 2022, 24, 737–740. [Google Scholar] [PubMed]
  2. Vilaca, T.; Eastell, R.; Schini, M. Osteoporosis in Men. Lancet Diabetes Endocrinol. 2022, 10, 273–283. [Google Scholar] [CrossRef] [PubMed]
  3. Compston, J. Reducing the Treatment Gap in Osteoporosis. Lancet Diabetes Endocrinol. 2020, 8, 7–9. [Google Scholar] [CrossRef] [PubMed]
  4. Compston, J.E.; McClung, M.R.; Leslie, W.D. Osteoporosis. Lancet 2019, 393, 364–376. [Google Scholar] [CrossRef] [PubMed]
  5. Hackshaw, K.V. The Search for Biomarkers in Fibromyalgia. Diagnostics 2021, 11, 156. [Google Scholar] [CrossRef] [PubMed]
  6. Siracusa, R.; Paola, R.D.; Cuzzocrea, S.; Impellizzeri, D. Fibromyalgia: Pathogenesis, Mechanisms, Diagnosis and Treatment Options Update. Int. J. Mol. Sci. 2021, 22, 3891. [Google Scholar] [CrossRef]
  7. Giorgi, V.; Sirotti, S.; Romano, M.E.; Marotto, D.; Ablin, J.N.; Salaffi, F.; Sarzi-Puttini, P. Fibromyalgia: One Year in Review 2022. Clin. Exp. Rheumatol. 2022, 40, 1065–1072. [Google Scholar] [CrossRef]
  8. Macfarlane, G.J.; Kronisch, C.; Dean, L.E.; Atzeni, F.; Häuser, W.; Fluß, E.; Choy, E.; Kosek, E.; Amris, K.; Branco, J.; et al. EULAR Revised Recommendations for the Management of Fibromyalgia. Ann. Rheum. Dis. 2017, 76, 318–328. [Google Scholar] [CrossRef]
  9. Pickering, G.; Achard, A.; Corriger, A.; Sickout-Arondo, S.; Macian, N.; Leray, V.; Lucchini, C.; Cardot, J.-M.; Pereira, B. Electrochemical Skin Conductance and Quantitative Sensory Testing on Fibromyalgia. Pain Pract. 2020, 20, 348–356. [Google Scholar] [CrossRef]
  10. Macian, N.; Pereira, B.; Shinjo, C.; Dubray, C.; Pickering, G. Fibromyalgia, Milnacipran and Experimental Pain Modulation: Study Protocol for a Double Blind Randomized Controlled Trial. Trials 2015, 16, 134. [Google Scholar] [CrossRef]
  11. Heidari, F.; Afshari, M.; Moosazadeh, M. Prevalence of Fibromyalgia in General Population and Patients, a Systematic Review and Meta-Analysis. Rheumatol. Int. 2017, 37, 1527–1539. [Google Scholar] [CrossRef] [PubMed]
  12. Wolfe, F.; Clauw, D.J.; Fitzcharles, M.-A.; Goldenberg, D.L.; Häuser, W.; Katz, R.L.; Mease, P.J.; Russell, A.S.; Russell, I.J.; Walitt, B. 2016 Revisions to the 2010/2011 Fibromyalgia Diagnostic Criteria. Semin. Arthritis Rheum. 2016, 46, 319–329. [Google Scholar] [CrossRef] [PubMed]
  13. Appelboom, T.; Schoutens, A. High Bone Turnover in Fibromyalgia. Calcif. Tissue Int. 1990, 46, 314–317. [Google Scholar] [CrossRef] [PubMed]
  14. Swezey, R.L.; Adams, J. Fibromyalgia: A Risk Factor for Osteoporosis. J. Rheumatol. 1999, 26, 2642–2644. [Google Scholar] [PubMed]
  15. Upala, S.; Yong, W.C.; Sanguankeo, A. Bone Mineral Density Is Decreased in Fibromyalgia Syndrome: A Systematic Review and Meta-Analysis. Rheumatol. Int. 2017, 37, 617–622. [Google Scholar] [CrossRef]
  16. Mateos Chapparo, F. Fibromyalgia and Osteoporosis. In Revista de Osteoporosis Y Metabolismo Mineral; Publicación Oficial SEIOMM: Madrid, Spain, 2011. [Google Scholar]
  17. Lee, Y.H.; Song, G.G. Association between Low Bone Mineral Density and Fibromyalgia: A Meta-Analysis. Clin. Rheumatol. 2017, 36, 2573–2579. [Google Scholar] [CrossRef]
  18. D’Agnelli, S.; Arendt-Nielsen, L.; Gerra, M.C.; Zatorri, K.; Boggiani, L.; Baciarello, M.; Bignami, E. Fibromyalgia: Genetics and Epigenetics Insights May Provide the Basis for the Development of Diagnostic Biomarkers. Mol. Pain 2019, 15, 1744806918819944. [Google Scholar] [CrossRef]
  19. Pickering, M.-E.; Millet, M.; Rousseau, J.-C.; Croset, M.; Szulc, P.; Borel, O.; Sornay Rendu, E.; Chapurlat, R. Selected Serum MicroRNA, Abdominal Aortic Calcification and Risk of Osteoporotic Fracture. PLoS ONE 2019, 14, e0216947. [Google Scholar] [CrossRef]
  20. Ge, D.-W.; Wang, W.-W.; Chen, H.-T.; Yang, L.; Cao, X.-J. Functions of MicroRNAs in Osteoporosis. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4784–4789. [Google Scholar]
  21. He, L.; Hannon, G.J. MicroRNAs: Small RNAs with a Big Role in Gene Regulation. Nat. Rev. Genet. 2004, 5, 522–531. [Google Scholar] [CrossRef]
  22. Feng, Q.; Zheng, S.; Zheng, J. The Emerging Role of MicroRNAs in Bone Remodeling and Its Therapeutic Implications for Osteoporosis. Biosci. Rep. 2018, 38, BSR20180453. [Google Scholar] [CrossRef]
  23. Yavropoulou, M.P.; Anastasilakis, A.D.; Makras, P.; Tsalikakis, D.G.; Grammatiki, M.; Yovos, J.G. Expression of MicroRNAs That Regulate Bone Turnover in the Serum of Postmenopausal Women with Low Bone Mass and Vertebral Fractures. Eur. J. Endocrinol. 2017, 176, 169–176. [Google Scholar] [CrossRef]
  24. Ko, N.-Y.; Chen, L.-R.; Chen, K.-H. The Role of Micro RNA and Long-Non-Coding RNA in Osteoporosis. Int. J. Mol. Sci. 2020, 21, 4886. [Google Scholar] [CrossRef]
  25. Yang, Y.; Yujiao, W.; Fang, W.; Linhui, Y.; Ziqi, G.; Zhichen, W.; Zirui, W.; Shengwang, W. The Roles of MiRNA, LncRNA and CircRNA in the Development of Osteoporosis. Biol. Res. 2020, 53, 40. [Google Scholar] [CrossRef] [PubMed]
  26. Jones, T.L.; Esa, M.S.; Li, K.H.C.; Krishnan, S.R.G.; Elgallab, G.M.; Pearce, M.S.; Young, D.A.; Birrell, F.N. Osteoporosis, Fracture, Osteoarthritis & Sarcopenia: A Systematic Review of Circulating MicroRNA Association. Bone 2021, 152, 116068. [Google Scholar] [CrossRef]
  27. 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] [PubMed]
  28. 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. 2017, 54, 7129–7136. [Google Scholar] [CrossRef] [PubMed]
  29. 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. 2015, 35, 635–642. [Google Scholar] [CrossRef]
  30. Erbacher, C.; Vaknine, S.; Moshitzky, G.; Lobentanzer, S.; Eisenberg, L.; Evdokimov, D.; Sommer, C.; Greenberg, D.S.; Soreq, H.; Üçeyler, N. Distinct CholinomiR Blood Cell Signature as a Potential Modulator of the Cholinergic System in Women with Fibromyalgia Syndrome. Cells 2022, 11, 1276. [Google Scholar] [CrossRef]
  31. Bjersing, J.L.; Lundborg, C.; Bokarewa, M.I.; Mannerkorpi, K. Profile of Cerebrospinal MicroRNAs in Fibromyalgia. PLoS ONE 2013, 8, e78762. [Google Scholar] [CrossRef]
  32. Clos-Garcia, M.; Andrés-Marin, N.; Fernández-Eulate, G.; Abecia, L.; Lavín, J.L.; van Liempd, S.; Cabrera, D.; Royo, F.; Valero, A.; Errazquin, N.; et al. Gut Microbiome and Serum Metabolome Analyses Identify Molecular Biomarkers and Altered Glutamate Metabolism in Fibromyalgia. eBio Med. 2019, 46, 499–511. [Google Scholar] [CrossRef]
  33. 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]
  34. 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] [PubMed]
  35. 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] [PubMed]
  36. Gu, H.; Shi, S.; Xiao, F.; Huang, Z.; Xu, J.; Chen, G.; Zhou, K.; Lu, L.; Yin, X. MiR-1-3p Regulates the Differentiation of Mesenchymal Stem Cells to Prevent Osteoporosis by Targeting Secreted Frizzled-Related Protein 1. Bone 2020, 137, 115444. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, R.; Lu, A.; Liu, W.; Yue, J.; Sun, Q.; Chen, J.; Luan, H.; Zhai, Y.; Li, B.; Jiang, Z.; et al. Searching for Valuable Differentially Expressed MiRNAs in Postmenopausal Osteoporosis by RNA Sequencing. J. Obstet. Gynaecol. Res. 2020, 46, 1183–1192. [Google Scholar] [CrossRef]
  38. Shao, J.-L.; Li, H.; Zhang, X.-R.; Zhang, X.; Li, Z.-Z.; Jiao, G.-L.; Sun, G.-D. Identification of Serum Exosomal MicroRNA Expression Profiling in Menopausal Females with Osteoporosis by High-Throughput Sequencing. Curr. Med. Sci. 2020, 40, 1161–1169. [Google Scholar] [CrossRef] [PubMed]
  39. Zhang, H.-G.; Wang, X.-B.; Zhao, H.; Zhou, C.-N. MicroRNA-9-5p Promotes Osteoporosis Development through Inhibiting Osteogenesis and Promoting Adipogenesis via Targeting Wnt3a. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 456–463. [Google Scholar] [CrossRef]
  40. Zhao, J.; Liu, S.; Zhang, W.; Ni, L.; Hu, Z.; Sheng, Z.; Yin, B. MiR-128 Inhibits the Osteogenic Differentiation in Osteoporosis by down-Regulating SIRT6 Expression. Biosci. Rep. 2019, 39, BSR20191405. [Google Scholar] [CrossRef]
  41. Chen, R.; Liao, X.; Chen, F.; Wang, B.; Huang, J.; Jian, G.; Huang, Z.; Yin, G.; Liu, H.; Jin, D. Circulating MicroRNAs, MiR-10b-5p, MiR-328-3p, MiR-100 and Let-7, Are Associated with Osteoblast Differentiation in Osteoporosis. Int. J. Clin. Exp. Pathol. 2018, 11, 1383–1390. [Google Scholar]
  42. Kocijan, R.; Muschitz, C.; Geiger, E.; Skalicky, S.; Baierl, A.; Dormann, R.; Plachel, F.; Feichtinger, X.; Heimel, P.; Fahrleitner-Pammer, A.; et al. Circulating MicroRNA Signatures in Patients With Idiopathic and Postmenopausal Osteoporosis and Fragility Fractures. J. Clin. Endocrinol. Metab. 2016, 101, 4125–4134. [Google Scholar] [CrossRef] [PubMed]
  43. Zarecki, P.; Hackl, M.; Grillari, J.; Debono, M.; Eastell, R. Serum MicroRNAs as Novel Biomarkers for Osteoporotic Vertebral Fractures. Bone 2020, 130, 115105. [Google Scholar] [CrossRef] [PubMed]
  44. Ramírez-Salazar, E.G.; Carrillo-Patiño, S.; Hidalgo-Bravo, A.; Rivera-Paredez, B.; Quiterio, M.; Ramírez-Palacios, P.; Patiño, N.; Valdés-Flores, M.; Salmerón, J.; Velázquez-Cruz, R. Serum MiRNAs MiR-140-3p and MiR-23b-3p as Potential Biomarkers for Osteoporosis and Osteoporotic Fracture in Postmenopausal Mexican-Mestizo Women. Gene 2018, 679, 19–27. [Google Scholar] [CrossRef] [PubMed]
  45. Teng, Z.; Zhu, Y.; Hao, Q.; Yu, X.; Teng, Y.; Yue, Q.; Zhang, X.; Lu, S. Long Non-Coding RNA Taurine Upregulated Gene 1 Is Downregulated in Osteoporosis and Influences the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. PeerJ 2021, 9, e11251. [Google Scholar] [CrossRef]
  46. Liu, H.; Liu, Q.; Wu, X.-P.; He, H.-B.; Fu, L. MiR-96 Regulates Bone Metabolism by Targeting Osterix. Clin. Exp. Pharmacol. Physiol. 2018, 45, 602–613. [Google Scholar] [CrossRef]
  47. Gao, Y.; Xiao, F.; Wang, C.; Wang, C.; Cui, P.; Zhang, X.; Chen, X. Long Noncoding RNA MALAT1 Promotes Osterix Expression to Regulate Osteogenic Differentiation by Targeting MiRNA-143 in Human Bone Marrow-Derived Mesenchymal Stem Cells. J. Cell. Biochem. 2018, 119, 6986–6996. [Google Scholar] [CrossRef]
  48. Fu, Y.; Xu, Y.; Chen, S.; Ouyang, Y.; Sun, G. MiR-151a-3p Promotes Postmenopausal Osteoporosis by Targeting SOCS5 and Activating JAK2/STAT3 Signaling. Rejuvenation Res 2020, 23, 313–323. [Google Scholar] [CrossRef]
  49. He, Y.; Chen, D.; Guo, Q.; Shi, P.; You, C.; Feng, Y. MicroRNA-151a-3p Functions in the Regulation of Osteoclast Differentiation: Significance to Postmenopausal Osteoporosis. Clin. Interv. Aging 2021, 16, 1357–1366. [Google Scholar] [CrossRef]
  50. Lin, C.; Yu, S.; Jin, R.; Xiao, Y.; Pan, M.; Pei, F.; Zhu, X.; Huang, H.; Zhang, Z.; Chen, S.; et al. Circulating MiR-338 Cluster Activities on Osteoblast Differentiation: Potential Diagnostic and Therapeutic Targets for Postmenopausal Osteoporosis. Theranostics 2019, 9, 3780–3797. [Google Scholar] [CrossRef]
  51. Kong, D.; Chen, T.; Zheng, X.; Yang, T.; Zhang, Y.; Shao, J. Comparative Profile of Exosomal MicroRNAs in Postmenopausal Women with Various Bone Mineral Densities by Small RNA Sequencing. Genomics 2021, 113, 1514–1521. [Google Scholar] [CrossRef]
  52. Chen, Z.; Bemben, M.G.; Bemben, D.A. Bone and Muscle Specific Circulating MicroRNAs in Postmenopausal Women Based on Osteoporosis and Sarcopenia Status. Bone 2019, 120, 271–278. [Google Scholar] [CrossRef] [PubMed]
  53. Ciuffi, S.; Marini, F.; Fossi, C.; Donati, S.; Giusti, F.; Botta, A.; Masi, L.; Isaia, G.; Marcocci, C.; Migliaccio, S.; et al. Circulating MicroRNAs as Biomarkers of Osteoporosis and Fragility Fractures. J. Clin. Endocrinol. Metab. 2022, 107, 2267–2285. [Google Scholar] [CrossRef] [PubMed]
  54. Cong, C.; Tian, J.; Gao, T.; Zhou, C.; Wang, Y.; Cui, X.; Zhu, L. LncRNA GAS5 Is Upregulated in Osteoporosis and Downregulates MiR-21 to Promote Apoptosis of Osteoclasts. Clin. Interv. Aging 2020, 15, 1163–1169. [Google Scholar] [CrossRef] [PubMed]
  55. Jiang, L.-B.; Tian, L.; Zhang, C.-G. Bone Marrow Stem Cells-Derived Exosomes Extracted from Osteoporosis Patients Inhibit Osteogenesis via MicroRNA-21/SMAD7. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 6221–6229. [Google Scholar] [CrossRef] [PubMed]
  56. Kelch, S.; Balmayor, E.R.; Seeliger, C.; Vester, H.; Kirschke, J.S.; van Griensven, M. MiRNAs in Bone Tissue Correlate to Bone Mineral Density and Circulating MiRNAs Are Gender Independent in Osteoporotic Patients. Sci. Rep. 2017, 7, 15861. [Google Scholar] [CrossRef]
  57. Li, H.; Wang, Z.; Fu, Q.; Zhang, J. Plasma MiRNA Levels Correlate with Sensitivity to Bone Mineral Density in Postmenopausal Osteoporosis Patients. Biomarkers 2014, 19, 553–556. [Google Scholar] [CrossRef]
  58. Panach, L.; Mifsut, D.; Tarín, J.J.; Cano, A.; García-Pérez, M.Á. Serum Circulating MicroRNAs as Biomarkers of Osteoporotic Fracture. Calcif. Tissue Int. 2015, 97, 495–505. [Google Scholar] [CrossRef]
  59. Seeliger, C.; Karpinski, K.; Haug, A.T.; Vester, H.; Schmitt, A.; Bauer, J.S.; van Griensven, M. Five Freely Circulating MiRNAs and Bone Tissue MiRNAs Are Associated with Osteoporotic Fractures. J. Bone Miner. Res. 2014, 29, 1718–1728. [Google Scholar] [CrossRef]
  60. Suarjana, I.N.; Isbagio, H.; Soewondo, P.; Rachman, I.A.; Sadikin, M.; Prihartono, J.; Malik, S.G.; Soeroso, J. The Role of Serum Expression Levels of Microrna-21 on Bone Mineral Density in Hypostrogenic Postmenopausal Women with Osteoporosis: Study on Level of RANKL, OPG, TGFβ-1, Sclerostin, RANKL/OPG Ratio, and Physical Activity. Acta Med. Indones 2019, 51, 245–252. [Google Scholar]
  61. Jiang, K.; Teng, G.-D.; Chen, Y.-Q. MicroRNA-23 Suppresses Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells by Targeting the MEF2C-Mediated MAPK Signaling Pathway. J. Gene Med. 2020, 22, e3216. [Google Scholar] [CrossRef]
  62. Li, Z.; Zhang, W.; Huang, Y. MiRNA-133a Is Involved in the Regulation of Postmenopausal Osteoporosis through Promoting Osteoclast Differentiation. Acta Biochim. Biophys. Sin. 2018, 50, 273–280. [Google Scholar] [CrossRef] [PubMed]
  63. Lv, H.; Sun, Y.; Zhang, Y. MiR-133 Is Involved in Estrogen Deficiency-Induced Osteoporosis through Modulating Osteogenic Differentiation of Mesenchymal Stem Cells. Med. Sci. Monit. 2015, 21, 1527–1534. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, Y.; Li, L.; Moore, B.T.; Peng, X.-H.; Fang, X.; Lappe, J.M.; Recker, R.R.; Xiao, P. MiR-133a in Human Circulating Monocytes: A Potential Biomarker Associated with Postmenopausal Osteoporosis. PLoS ONE 2012, 7, e34641. [Google Scholar] [CrossRef]
  65. Fu, Y.; Hu, X.; Gao, Y.; Li, K.; Fu, Q.; Liu, Q.; Liu, D.; Zhang, Z.; Qiao, J. LncRNA ROR/MiR-145-5p Axis Modulates the Osteoblasts Proliferation and Apoptosis in Osteoporosis. Bioengineered 2021, 12, 7714–7723. [Google Scholar] [CrossRef] [PubMed]
  66. Sun, M.; Hu, L.; Wang, S.; Huang, T.; Zhang, M.; Yang, M.; Zhen, W.; Yang, D.; Lu, W.; Guan, M.; et al. Circulating MicroRNA-19b Identified From Osteoporotic Vertebral Compression Fracture Patients Increases Bone Formation. J. Bone Miner. Res. 2020, 35, 306–316. [Google Scholar] [CrossRef]
  67. Al-Rawaf, H.A.; Alghadir, A.H.; Gabr, S.A. Circulating MicroRNA Expression, Vitamin D, and Hypercortisolism as Predictors of Osteoporosis in Elderly Postmenopausal Women. Dis. Markers 2021, 2021, 3719919. [Google Scholar] [CrossRef]
  68. Bedene, A.; Mencej Bedrač, S.; Ješe, L.; Marc, J.; Vrtačnik, P.; Preželj, J.; Kocjan, T.; Kranjc, T.; Ostanek, B. MiR-148a the Epigenetic Regulator of Bone Homeostasis Is Increased in Plasma of Osteoporotic Postmenopausal Women. Wien. Klin. Wochenschr. 2016, 128, 519–526. [Google Scholar] [CrossRef]
  69. De-Ugarte, L.; Yoskovitz, G.; Balcells, S.; Güerri-Fernández, R.; Martinez-Diaz, S.; Mellibovsky, L.; Urreizti, R.; Nogués, X.; Grinberg, D.; García-Giralt, N.; et al. MiRNA Profiling of Whole Trabecular Bone: Identification of Osteoporosis-Related Changes in MiRNAs in Human Hip Bones. BMC Med. Genom. 2015, 8, 75. [Google Scholar] [CrossRef]
  70. Wang, C.-G.; Hu, Y.-H.; Su, S.-L.; Zhong, D. LncRNA DANCR and MiR-320a Suppressed Osteogenic Differentiation in Osteoporosis by Directly Inhibiting the Wnt/β-Catenin Signaling Pathway. Exp. Mol. Med. 2020, 52, 1310–1325. [Google Scholar] [CrossRef]
  71. Guo, X.; Wei, S.; Xu, F.; Cai, X.; Wang, H.; Ding, R. MicroRNA-532-5p Is Implicated in the Regulation of Osteoporosis by Forkhead Box O1 and Osteoblast Differentiation. BMC Musculoskelet. Disord. 2020, 21, 296. [Google Scholar] [CrossRef]
  72. Li, T.; Jiang, H.; Li, Y.; Zhao, X.; Ding, H. Estrogen Promotes LncRNA H19 Expression to Regulate Osteogenic Differentiation of BMSCs and Reduce Osteoporosis via MiR-532-3p/SIRT1 Axis. Mol. Cell. Endocrinol. 2021, 527, 111171. [Google Scholar] [CrossRef]
  73. Zhou, Z.; Lu, Y.; Wang, Y.; Du, L.; Zhang, Y.; Tao, J. Let-7c Regulates Proliferation and Osteodifferentiation of Human Adipose-Derived Mesenchymal Stem Cells under Oxidative Stress by Targeting SCD-1. Am. J. Physiol. Cell Physiol. 2019, 316, C57–C69. [Google Scholar] [CrossRef]
  74. Weilner, S.; Skalicky, S.; Salzer, B.; Keider, V.; Wagner, M.; Hildner, F.; Gabriel, C.; Dovjak, P.; Pietschmann, P.; Grillari-Voglauer, R.; et al. Differentially Circulating MiRNAs after Recent Osteoporotic Fractures Can Influence Osteogenic Differentiation. Bone 2015, 79, 43–51. [Google Scholar] [CrossRef]
  75. Jin, D.; Wu, X.; Yu, H.; Jiang, L.; Zhou, P.; Yao, X.; Meng, J.; Wang, L.; Zhang, M.; Zhang, Y. Systematic Analysis of LncRNAs, MRNAs, CircRNAs and MiRNAs in Patients with Postmenopausal Osteoporosis. Am. J. Transl. Res. 2018, 10, 1498–1510. [Google Scholar] [PubMed]
  76. Wu, W.; Li, Q.; Liu, Y.-F.; Li, Y. LncRNA GAS5 Regulates Angiogenesis by Targeting MiR-10a-3p/VEGFA in Osteoporosis. Mol. Med. Rep. 2021, 24, 711. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, Q.; Miao, Y.; Qian, Z.; Chen, L.; Lu, T.; Xu, Y.; Jiang, X.; Shen, Y. MicroRNA-15a-5p Plays a Role in Osteogenic MC3T3-E1 Cells Differentiation by Targeting PDCD4 (Programmed Cell Death 4) via Wnt/β-Catenin Dependent Signaling Pathway. Bioengineered 2021, 12, 8173–8185. [Google Scholar] [CrossRef] [PubMed]
  78. Feng, Y.; Ding, L.; Li, L. LPS-Inducible CircAtp9b Is Highly Expressed in Osteoporosis and Promotes the Apoptosis of Osteoblasts by Reducing the Formation of Mature MiR-17-92a. J. Orthop. Surg. Res. 2022, 17, 193. [Google Scholar] [CrossRef]
  79. Wang, L.; Dong, J.; Ma, J.; Lu, Q.; Shan, B.; Han, T.; Xie, P.; Zuo, X. By Inhibiting ADCY5, MiR-18a-3p Promotes Osteoporosis and Possibly Contributes to Spinal Fracture. Biochem. Biophys. Res. Commun. 2021, 550, 49–55. [Google Scholar] [CrossRef]
  80. Zhao, M.; Dong, J.; Liao, Y.; Lu, G.; Pan, W.; Zhou, H.; Zuo, X.; Shan, B. MicroRNA MiR-18a-3p Promotes Osteoporosis and Possibly Contributes to Spinal Fracture by Inhibiting the Glutamate AMPA Receptor Subunit 1 Gene (GRIA1). Bioengineered 2022, 13, 370–382. [Google Scholar] [CrossRef]
  81. Chen, R.; Qiu, H.; Tong, Y.; Liao, F.; Hu, X.; Qiu, Y.; Liao, Y. MiRNA-19a-3p Alleviates the Progression of Osteoporosis by Targeting HDAC4 to Promote the Osteogenic Differentiation of HMSCs. Biochem. Biophys. Res. Commun. 2019, 516, 666–672. [Google Scholar] [CrossRef]
  82. Xiaoling, G.; Shuaibin, L.; Kailu, L. MicroRNA-19b-3p Promotes Cell Proliferation and Osteogenic Differentiation of BMSCs by Interacting with LncRNA H19. BMC Med. Genet. 2020, 21, 11. [Google Scholar] [CrossRef]
  83. Jia, X.; Yang, M.; Hu, W.; Cai, S. Overexpression of MiRNA-22-3p Attenuates Osteoporosis by Targeting MAPK14. Exp. Ther. Med. 2021, 22, 692. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, C.; He, H.; Wang, L.; Jiang, Y.; Xu, Y. Reduced MiR-144-3p Expression in Serum and Bone Mediates Osteoporosis Pathogenesis by Targeting RANK. Biochem. Cell Biol. 2018, 96, 627–635. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, D.; Li, Z.; Cao, J.; Shen, F.; Wei, G. MicroRNA-25-3p Suppresses Osteogenic Differentiation of BMSCs in Patients with Osteoporosis by Targeting ITGB3. Acta Histochem. 2022, 124, 151926. [Google Scholar] [CrossRef] [PubMed]
  86. Yuan, F.; Chen, H.; Hu, P.; Su, P.; Guan, X. MiR-26a Regulates the Expression of Serum IGF-1 in Patients with Osteoporosis and Its Effect on Proliferation and Apoptosis of Mouse Chondrocytes. J. Musculoskelet. Neuronal Interact. 2021, 21, 298–307. [Google Scholar]
  87. Fu, Y.-C.; Zhao, S.-R.; Zhu, B.-H.; Guo, S.-S.; Wang, X.-X. MiRNA-27a-3p Promotes Osteogenic Differentiation of Human Mesenchymal Stem Cells through Targeting ATF3. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 73–80. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, Y.; Zhou, X.; Wang, D. Mesenchymal Stem Cell-Derived Extracellular Vesicles Inhibit Osteoporosis via MicroRNA-27a-Induced Inhibition of DKK2-Mediated Wnt/β-Catenin Pathway. Inflammation 2022, 45, 780–799. [Google Scholar] [CrossRef]
  89. Xu, Y.; Li, D.; Zhu, Z.; Li, L.; Jin, Y.; Ma, C.; Zhang, W. MiR-27a-3p Negatively Regulates Osteogenic Differentiation of MC3T3-E1 Preosteoblasts by Targeting Osterix. Mol. Med. Rep. 2020, 22, 1717–1726. [Google Scholar] [CrossRef] [PubMed]
  90. You, L.; Pan, L.; Chen, L.; Gu, W.; Chen, J. MiR-27a Is Essential for the Shift from Osteogenic Differentiation to Adipogenic Differentiation of Mesenchymal Stem Cells in Postmenopausal Osteoporosis. Cell. Physiol. Biochem. 2016, 39, 253–265. [Google Scholar] [CrossRef] [PubMed]
  91. Yu, J.; Xiao, M.; Ren, G. Long Non-Coding RNA XIST Promotes Osteoporosis by Inhibiting the Differentiation of Bone Marrow Mesenchymal Stem Cell by Sponging MiR-29b-3p That Suppresses Nicotinamide N-Methyltransferase. Bioengineered 2021, 12, 6057–6069. [Google Scholar] [CrossRef]
  92. Zhang, X.; Wang, W.; Wang, Y.; Zhao, H.; Han, X.; Zhao, T.; Qu, P. Extracellular Vesicle-Encapsulated MiR-29b-3p Released From Bone Marrow-Derived Mesenchymal Stem Cells Underpins Osteogenic Differentiation. Front. Cell Dev. Biol. 2020, 8, 581545. [Google Scholar] [CrossRef]
  93. Che, M.; Gong, W.; Zhao, Y.; Liu, M. Long Noncoding RNA HCG18 Inhibits the Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells in Osteoporosis by Targeting MiR-30a-5p/NOTCH1 Axis. Mol. Med. 2020, 26, 106. [Google Scholar] [CrossRef]
  94. Chen, H.; Jiang, H.; Can, D.; Xu, H.; Zhang, K.; Guo, S. Evaluation of MicroRNA 125b as a Potential Biomarker for Postmenopausal Osteoporosis. Trop. J. Pharm. Res. 2017, 16, 641–647. [Google Scholar] [CrossRef]
  95. Zhang, H.-L.; Du, X.-Y.; Dong, Q.-R. LncRNA XIXT Promotes Osteogenic Differentiation of Bone Mesenchymal Stem Cells and Alleviates Osteoporosis Progression by Targeting MiRNA-30a-5p. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8721–8729. [Google Scholar] [CrossRef]
  96. Weilner, S.; Schraml, E.; Wieser, M.; Messner, P.; Schneider, K.; Wassermann, K.; Micutkova, L.; Fortschegger, K.; Maier, A.B.; Westendorp, R.; et al. Secreted Microvesicular MiR-31 Inhibits Osteogenic Differentiation of Mesenchymal Stem Cells. Aging Cell 2016, 15, 744–754. [Google Scholar] [CrossRef] [PubMed]
  97. Qiao, L.; Li, C.-G.; Liu, D. CircRNA_0048211 Protects Postmenopausal Osteoporosis through Targeting MiRNA-93-5p to Regulate BMP2. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3459–3466. [Google Scholar] [CrossRef] [PubMed]
  98. Ding, W.; Ding, S.; Li, J.; Peng, Z.; Hu, P.; Zhang, T.; Pan, L. Aberrant Expression of MiR-100 in Plasma of Patients with Osteoporosis and Its Potential Diagnostic Value. Clin. Lab. 2019, 65, 1727–1733. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, R.; Zhang, M.; Hu, Y.; He, J.; Lin, Q.; Peng, N. MiR-100-5p Inhibits Osteogenic Differentiation of Human Bone Mesenchymal Stromal Cells by Targeting TMEM135. Hum. Cell 2022, 35, 1671–1683. [Google Scholar] [CrossRef] [PubMed]
  100. Mandourah, A.Y.; Ranganath, L.; Barraclough, R.; Vinjamuri, S.; Hof, R.V.; Hamill, S.; Czanner, G.; Dera, A.A.; Wang, D.; Barraclough, D.L. Circulating MicroRNAs as Potential Diagnostic Biomarkers for Osteoporosis. Sci. Rep. 2018, 8, 8421. [Google Scholar] [CrossRef] [PubMed]
  101. Chen, S.; Yang, L.; Jie, Q.; Lin, Y.-S.; Meng, G.-L.; Fan, J.-Z.; Zhang, J.-K.; Fan, J.; Luo, Z.-J.; Liu, J. MicroRNA-125b Suppresses the Proliferation and Osteogenic Differentiation of Human Bone Marrow-derived Mesenchymal Stem Cells. Mol. Med. Rep. 2014, 9, 1820–1826. [Google Scholar] [CrossRef]
  102. Wang, G.; Zhang, L.; Yan, C.; Wang, F.; Zhang, Y. Overexpression of MiR125b Promotes Osteoporosis Through MiR-125b-TRAF6 Pathway in Postmenopausal Ovariectomized Rats. Diabetes Metab. Syndr. Obes. 2021, 14, 671–682. [Google Scholar] [CrossRef]
  103. Zhang, D.-W.; Chen, T.; Li, J.-X.; Wang, H.-G.; Huang, Z.-W.; Lv, H. Circ_0134944 Inhibits Osteogenesis through MiR-127-5p/PDX1/SPHK1 Pathway. Regen. Ther. 2021, 18, 391–400. [Google Scholar] [CrossRef] [PubMed]
  104. Shi, X.; Zhang, Z. MicroRNA-135a-5p Is Involved in Osteoporosis Progression through Regulation of Osteogenic Differentiation by Targeting RUNX2. Exp. Ther. Med. 2019, 18, 2393–2400. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, B.; Yang, W.; Zhao, H.; Liu, K.; Deng, A.; Zhang, G.; Pan, K. Abnormal Expression of MiR-135b-5p in Bone Tissue of Patients with Osteoporosis and Its Role and Mechanism in Osteoporosis Progression. Exp. Ther. Med. 2020, 19, 1042–1050. [Google Scholar] [CrossRef] [PubMed]
  106. Yu, Y.; Yao, P.; Wang, Z.; Xie, W. Down-Regulation of FTX Promotes the Differentiation of Osteoclasts in Osteoporosis through the Notch1 Signaling Pathway by Targeting MiR-137. BMC Musculoskelet. Disord. 2020, 21, 456. [Google Scholar] [CrossRef]
  107. Yin, R.; Jiang, J.; Deng, H.; Wang, Z.; Gu, R.; Wang, F. MiR-140-3p Aggregates Osteoporosis by Targeting PTEN and Activating PTEN/PI3K/AKT Signaling Pathway. Hum. Cell 2020, 33, 569–581. [Google Scholar] [CrossRef] [PubMed]
  108. Tang, L.; Lu, W.; Huang, J.; Tang, X.; Zhang, H.; Liu, S. MiR-144 Promotes the Proliferation and Differentiation of Bone Mesenchymal Stem Cells by Downregulating the Expression of SFRP1. Mol. Med. Rep. 2019, 20, 270–280. [Google Scholar] [CrossRef] [PubMed]
  109. Zhao, S.-L.; Wen, Z.-X.; Mo, X.-Y.; Zhang, X.-Y.; Li, H.-N.; Cheung, W.-H.; Fu, D.; Zhang, S.-H.; Wan, Y.; Chen, B.-L. Bone-Metabolism-Related Serum MicroRNAs to Diagnose Osteoporosis in Middle-Aged and Elderly Women. Diagnostics 2022, 12, 2872. [Google Scholar] [CrossRef]
  110. Nakashima, H.; Ando, K.; Kobayashi, K.; Seki, T.; Ishizuka, S.; Fujii, R.; Takegami, Y.; Yamada, H.; Ando, Y.; Suzuki, K.; et al. Associations of Serum MicroRNA with Bone Mineral Density in Community-Dwelling Subjects: The Yakumo Study. Biomed. Res. Int. 2020, 2020, 5047243. [Google Scholar] [CrossRef]
  111. Wang, F.; Deng, H.; Chen, J.; Wang, Z.; Yin, R. LncRNA MIAT Can Regulate the Proliferation, Apoptosis, and Osteogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells by Targeting MiR-150-5p. Bioengineered 2022, 13, 6343–6352. [Google Scholar] [CrossRef]
  112. Ismail, S.M.; El Boghdady, N.A.; Hamoud, H.S.; Shabayek, M.I. Evaluation of Circulating MiRNA-208a-3p, MiRNA-155-5p and MiRNA-637 as Potential Non-Invasive Biomarkers and the Possible Mechanistic Insights into Pre- and Postmenopausal Osteoporotic Females. Arch. Biochem. Biophys. 2020, 684, 108331. [Google Scholar] [CrossRef]
  113. Ming, Y.; Liu, Z.-P. Overexpression of LncRNA-NEF Regulates the MiR-155/PTEN Axis to Inhibit Adipogenesis and Promote Osteogenesis. Kaohsiung J. Med. Sci. 2021, 37, 930–939. [Google Scholar] [CrossRef] [PubMed]
  114. Pertusa, C.; Tarín, J.J.; Cano, A.; García-Pérez, M.Á.; Mifsut, D. Serum MicroRNAs in Osteoporotic Fracture and Osteoarthritis: A Genetic and Functional Study. Sci. Rep. 2021, 11, 19372. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, J.; Lin, X.; Chen, C.; Li, S.; Zhang, S.; Chen, Z.; Li, D.; Zhao, F.; Yang, C.; Yin, C.; et al. Circulating MiR-181c-5p and MiR-497-5p Are Potential Biomarkers for Prognosis and Diagnosis of Osteoporosis. J. Clin. Endocrinol. Metab. 2020, 105, dgz300. [Google Scholar] [CrossRef] [PubMed]
  116. Garmilla-Ezquerra, P.; Sañudo, C.; Delgado-Calle, J.; Pérez-Nuñez, M.I.; Sumillera, M.; Riancho, J.A. Analysis of the Bone MicroRNome in Osteoporotic Fractures. Calcif. Tissue Int. 2015, 96, 30–37. [Google Scholar] [CrossRef] [PubMed]
  117. Meng, J.; Zhang, D.; Pan, N.; Sun, N.; Wang, Q.; Fan, J.; Zhou, P.; Zhu, W.; Jiang, L. Identification of MiR-194-5p as a Potential Biomarker for Postmenopausal Osteoporosis. PeerJ 2015, 3, e971. [Google Scholar] [CrossRef] [PubMed]
  118. Lv, R.; Pan, X.; Song, L.; Sun, Q.; Guo, C.; Zou, S.; Zhou, Q. MicroRNA-200a-3p Accelerates the Progression of Osteoporosis by Targeting Glutaminase to Inhibit Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Biomed. Pharmacother. 2019, 116, 108960. [Google Scholar] [CrossRef]
  119. Qiao, L.; Liu, D.; Li, C.-G.; Wang, Y.-J. MiR-203 Is Essential for the Shift from Osteogenic Differentiation to Adipogenic Differentiation of Mesenchymal Stem Cells in Postmenopausal Osteoporosis. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5804–5814. [Google Scholar] [CrossRef] [PubMed]
  120. Xia, Z.-L.; Wang, Y.; Sun, Q.-D.; Du, X.-F. MiR-203 Is Involved in Osteoporosis by Regulating DKK1 and Inhibiting Osteogenic Differentiation of MSCs. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5098–5105. [Google Scholar] [CrossRef]
  121. Huang, M.; Li, X.; Zhou, C.; Si, M.; Zheng, H.; Chen, L.; Ding, H. Noncoding RNA MiR-205-5p Mediates Osteoporosis Pathogenesis and Osteoblast Differentiation by Regulating RUNX2. J. Cell. Biochem. 2020, 121, 4196–4203. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, J.-J.; Peng, W.-X.; Zhang, M.-B. LncRNA KCNQ1OT1 Promotes Osteogenic Differentiation via MiR-205-5p/RICTOR Axis. Exp. Cell Res. 2022, 415, 113119. [Google Scholar] [CrossRef] [PubMed]
  123. Lu, Z.; Wang, D.; Wang, X.; Zou, J.; Sun, J.; Bi, Z. MiR-206 Regulates the Progression of Osteoporosis via Targeting HDAC4. Eur. J. Med. Res. 2021, 26, 8. [Google Scholar] [CrossRef] [PubMed]
  124. Hu, M.; Zhu, X.; Yuan, H.; Li, H.; Liao, H.; Chen, S. The Function and Mechanism of the MiR-210-3p/KRAS Axis in Bone Marrow-Derived Mesenchymal Stem Cell from Patients with Osteoporosis. J. Tissue Eng. Regen. Med. 2021, 15, 699–711. [Google Scholar] [CrossRef]
  125. Wang, H.; Shi, X.; Guo, Z.; Zhao, F.; He, W.; Kang, M.; Lv, Z. MicroRNA-211-5p Predicts the Progression of Postmenopausal Osteoporosis and Attenuates Osteogenesis by Targeting Dual Specific Phosphatase 6. Bioengineered 2022, 13, 5709–5723. [Google Scholar] [CrossRef]
  126. Mohamad, N.; Nabih, E.S.; Zakaria, Z.M.; Nagaty, M.M.; Metwaly, R.G. Insight into the Possible Role of MiR-214 in Primary Osteoporosis via Osterix. J. Cell. Biochem. 2019, 120, 15518–15526. [Google Scholar] [CrossRef]
  127. Yin, Z.; Shen, J.; Wang, Q.; Wen, L.; Qu, W.; Zhang, Y. MiR-215-5p Regulates Osteoporosis Development and Osteogenic Differentiation by Targeting XIAP. BMC Musculoskelet. Disord. 2022, 23, 789. [Google Scholar] [CrossRef]
  128. Gao, G.-C.; Yang, D.-W.; Liu, W. LncRNA TERC Alleviates the Progression of Osteoporosis by Absorbing MiRNA-217 to Upregulate RUNX2. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 526–534. [Google Scholar] [CrossRef]
  129. Yang, L.; Zeng, Z.; Kang, N.; Yang, J.-C.; Wei, X.; Hai, Y. Circ-VANGL1 Promotes the Progression of Osteoporosis by Absorbing MiRNA-217 to Regulate RUNX2 Expression. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 949–957. [Google Scholar] [CrossRef]
  130. Guo, M.; Liu, N.; Guo, Z. MiR-221-5p/Smad3 Axis in Osteoclastogenesis and Its Function: Potential Therapeutic Target for Osteoporosis. Steroids 2022, 185, 109063. [Google Scholar] [CrossRef] [PubMed]
  131. Zhang, Y.; Gao, Y.; Cai, L.; Li, F.; Lou, Y.; Xu, N.; Kang, Y.; Yang, H. MicroRNA-221 Is Involved in the Regulation of Osteoporosis through Regulates RUNX2 Protein Expression and Osteoblast Differentiation. Am. J. Transl. Res. 2017, 9, 126–135. [Google Scholar]
  132. Wang, Z.; Ge, X.; Wang, Y.; Liang, Y.; Shi, H.; Zhao, T. Mechanism of Dexmedetomidine Regulating Osteogenesis-Angiogenesis Coupling through the MiR-361-5p/VEGFA Axis in Postmenopausal Osteoporosis. Life Sci. 2021, 275, 119273. [Google Scholar] [CrossRef]
  133. Li, M.; Luo, R.; Yang, W.; Zhou, Z.; Li, C. MiR-363-3p Is Activated by MYB and Regulates Osteoporosis Pathogenesis via PTEN/PI3K/AKT Signaling Pathway. Vitr. Cell. Dev. Biol. Anim. 2019, 55, 376–386. [Google Scholar] [CrossRef]
  134. Cheng, F.; Yang, M.-M.; Yang, R.-H. MiRNA-365a-3p Promotes the Progression of Osteoporosis by Inhibiting Osteogenic Differentiation via Targeting RUNX2. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7766–7774. [Google Scholar] [CrossRef]
  135. Li, L.-Y.; Wang, X.-L.; Wang, G.-S.; Zhao, H.-Y. MiR-373 Promotes the Osteogenic Differentiation of BMSCs from the Estrogen Deficiency Induced Osteoporosis. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7247–7255. [Google Scholar] [CrossRef]
  136. Lei, N.-B.; Liang, X.; Wang, P.; Liu, Q.; Wang, W.-G. Teriparatide Alleviates Osteoporosis by Promoting Osteogenic Differentiation of HMSCs via MiR-375/RUNX2 Axis. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 11043–11050. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, W.; Li, T.; Feng, S. Knockdown of Long Non-Coding RNA HOTAIR Promotes Bone Marrow Mesenchymal Stem Cell Differentiation by Sponging MicroRNA MiR-378g That Inhibits Nicotinamide N-Methyltransferase. Bioengineered 2021, 12, 12482–12497. [Google Scholar] [CrossRef]
  138. Zhang, H.; Ding, W.; Ji, F.; Wu, D. MicroRNA-410 Participates in the Pathological Process of Postmenopausal Osteoporosis by Downregulating Bone Morphogenetic Protein-2. Exp. Ther. Med. 2019, 18, 3659–3666. [Google Scholar] [CrossRef]
  139. Cao, Z.; Moore, B.T.; Wang, Y.; Peng, X.-H.; Lappe, J.M.; Recker, R.R.; Xiao, P. MiR-422a as a Potential Cellular MicroRNA Biomarker for Postmenopausal Osteoporosis. PLoS ONE 2014, 9, e97098. [Google Scholar] [CrossRef]
  140. Ma, H.; Li, M.; Jia, Z.; Chen, X.; Bu, N. MicroRNA-455-3p Promotes Osteoblast Differentiation via Targeting HDAC2. Injury 2022, 53, 3636–3641. [Google Scholar] [CrossRef]
  141. Li, K.; Chen, S.; Cai, P.; Chen, K.; Li, L.; Yang, X.; Yi, J.; Luo, X.; Du, Y.; Zheng, H. MiRNA-483-5p Is Involved in the Pathogenesis of Osteoporosis by Promoting Osteoclast Differentiation. Mol. Cell. Probes 2020, 49, 101479. [Google Scholar] [CrossRef]
  142. Zhao, F.; Xu, Y.; Ouyang, Y.; Wen, Z.; Zheng, G.; Wan, T.; Sun, G. Silencing of MiR-483-5p Alleviates Postmenopausal Osteoporosis by Targeting SATB2 and PI3K/AKT Pathway. Aging 2021, 13, 6945–6956. [Google Scholar] [CrossRef] [PubMed]
  143. Zhou, B.; Peng, K.; Wang, G.; Chen, W.; Liu, P.; Chen, F.; Kang, Y. MiR-483-3p Promotes the Osteogenesis of Human Osteoblasts by Targeting Dikkopf 2 (DKK2) and the Wnt Signaling Pathway. Int. J. Mol. Med. 2020, 46, 1571–1581. [Google Scholar] [CrossRef]
  144. Zhang, S.-Y.; Gao, F.; Peng, C.-G.; Zheng, C.-J.; Wu, M.-F. MiR-485-5p Promotes Osteoporosis via Targeting Osterix. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4792–4799. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, Z.; Jiang, W.; Hu, M.; Gao, R.; Zhou, X. MiR-486-3p Promotes Osteogenic Differentiation of BMSC by Targeting CTNNBIP1 and Activating the Wnt/β-Catenin Pathway. Biochem. Biophys. Res. Commun. 2021, 566, 59–66. [Google Scholar] [CrossRef] [PubMed]
  146. Zhao, H.; Yang, Y.; Wang, Y.; Feng, X.; Deng, A.; Ou, Z.; Chen, B. MicroRNA-497-5p Stimulates Osteoblast Differentiation through HMGA2-Mediated JNK Signaling Pathway. J. Orthop. Surg. Res. 2020, 15, 515. [Google Scholar] [CrossRef]
  147. Feng, J.; Wang, J.-X.; Li, C.-H. LncRNA GAS5 Overexpression Alleviates the Development of Osteoporosis through Promoting Osteogenic Differentiation of MSCs via Targeting MicroRNA-498 to Regulate RUNX2. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7757–7765. [Google Scholar] [CrossRef]
  148. Chen, C.; Cheng, P.; Xie, H.; Zhou, H.-D.; Wu, X.-P.; Liao, E.-Y.; Luo, X.-H. MiR-503 Regulates Osteoclastogenesis via Targeting RANK. J. Bone Miner. Res. 2014, 29, 338–347. [Google Scholar] [CrossRef]
  149. Ramírez-Salazar, E.G.; Almeraya, E.V.; López-Perez, T.V.; Patiño, N.; Salmeron, J.; Velázquez-Cruz, R. MicroRNA-548-3p Overexpression Inhibits Proliferation, Migration and Invasion in Osteoblast-like Cells by Targeting STAT1 and MAFB. J. Biochem. 2020, 168, 203–211. [Google Scholar] [CrossRef]
  150. Luo, B.; Yang, J.-F.; Wang, Y.-H.; Qu, G.-B.; Hao, P.-D.; Zeng, Z.-J.; Yuan, J.; Yang, R.; Yuan, Y. MicroRNA-579-3p Promotes the Progression of Osteoporosis by Inhibiting Osteogenic Differentiation of Mesenchymal Stem Cells through Regulating Sirt1. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6791–6799. [Google Scholar] [CrossRef]
  151. Yin, J.; Xiao, W.; Zhao, Q.; Sun, J.; Zhou, W.; Zhao, W. MicroRNA-582-3p Regulates Osteoporosis through Regulating Homeobox A10 and Osteoblast Differentiation. Immunopharmacol. Immunotoxicol. 2022, 44, 421–428. [Google Scholar] [CrossRef]
  152. Chen, S.; Dai, M. Lipopolysaccharide-Induced LncRNA TMC3-AS1 Is Highly Expressed in Osteoporosis and Promotes Osteoblast Apoptosis by Suppressing the Formation of Mature MiR-708. Int. J. Gen. Med. 2022, 15, 3345–3352. [Google Scholar] [CrossRef]
  153. De-La-Cruz-Montoya, A.H.; Ramírez-Salazar, E.G.; Martínez-Aguilar, M.M.; González-de-la-Rosa, P.M.; Quiterio, M.; Abreu-Goodger, C.; Salmerón, J.; Velázquez-Cruz, R. Identification of MiR-708-5p in Peripheral Blood Monocytes: Potential Marker for Postmenopausal Osteoporosis in Mexican-Mestizo Population. Exp. Biol. Med. 2018, 243, 1027–1036. [Google Scholar] [CrossRef] [PubMed]
  154. Mei, L.; Li, M.; Zhang, T. MicroRNA MiR-874-3p Inhibits Osteoporosis by Targeting Leptin (LEP). Bioengineered 2021, 12, 11756–11767. [Google Scholar] [CrossRef]
  155. Xu, G.; Ding, Z.; Shi, H.-F. The Mechanism of MiR-889 Regulates Osteogenesis in Human Bone Marrow Mesenchymal Stem Cells. J. Orthop. Surg. Res. 2019, 14, 366. [Google Scholar] [CrossRef] [PubMed]
  156. Zha, J.-P.; Wang, X.-Q.; Di, J. MiR-920 Promotes Osteogenic Differentiation of Human Bone Mesenchymal Stem Cells by Targeting HOXA7. J. Orthop. Surg. Res. 2020, 15, 254. [Google Scholar] [CrossRef]
  157. Yang, X.-M.; Song, Y.-Q.; Li, L.; Liu, D.-M.; Chen, G.-D. MiR-1249-5p Regulates the Osteogenic Differentiation of ADSCs by Targeting PDX1. J. Orthop. Surg. Res. 2021, 16, 10. [Google Scholar] [CrossRef] [PubMed]
  158. Jiménez-Ortega, R.F.; Ramírez-Salazar, E.G.; Parra-Torres, A.Y.; Muñoz-Montero, S.A.; Rangel-Escareňo, C.; Salido-Guadarrama, I.; Rodriguez-Dorantes, M.; Quiterio, M.; Salmerón, J.; Velázquez-Cruz, R. Identification of MicroRNAs in Human Circulating Monocytes of Postmenopausal Osteoporotic Mexican-Mestizo Women: A Pilot Study. Exp. Ther. Med. 2017, 14, 5464–5472. [Google Scholar]
  159. Yang, Q.; Zhou, Y.; Wang, T.; Cai, P.; Fu, W.; Wang, J.; Li, X. MiRNA-1271-5p Regulates Osteogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells by Targeting Forkhead Box O1 (FOXO1). Cell Biol. Int. 2021, 45, 1468–1476. [Google Scholar] [CrossRef]
  160. Zhou, J.-G.; Hua, Y.; Liu, S.-W.; Hu, W.-Q.; Qian, R.; Xiong, L. MicroRNA-1286 Inhibits Osteogenic Differentiation of Mesenchymal Stem Cells to Promote the Progression of Osteoporosis via Regulating FZD4 Expression. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1–10. [Google Scholar] [CrossRef]
  161. Wang, Q.; Wang, C.-H.; Meng, Y. MicroRNA-1297 Promotes the Progression of Osteoporosis through Regulation of Osteogenesis of Bone Marrow Mesenchymal Stem Cells by Targeting WNT5A. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4541–4550. [Google Scholar] [CrossRef]
  162. Du, M.; Wu, B.; Fan, S.; Liu, Y.; Ma, X.; Fu, X. SNHG14 Induces Osteogenic Differentiation of Human Stromal (Mesenchymal) Stem Cells in Vitro by Downregulating MiR-2861. BMC Musculoskelet. Disord. 2020, 21, 525. [Google Scholar] [CrossRef] [PubMed]
  163. De-Ugarte, L.; Caro-Molina, E.; Rodríguez-Sanz, M.; García-Pérez, M.A.; Olmos, J.M.; Sosa-Henríquez, M.; Pérez-Cano, R.; Gómez-Alonso, C.; Del Rio, L.; Mateo-Agudo, J.; et al. SNPs in Bone-Related MiRNAs Are Associated with the Osteoporotic Phenotype. Sci. Rep. 2017, 7, 516. [Google Scholar] [CrossRef]
  164. Huang, K.; Zhang, J.-X.; Han, L.; You, Y.-P.; Jiang, T.; Pu, P.-Y.; Kang, C.-S. MicroRNA Roles in Beta-Catenin Pathway. Mol. Cancer 2010, 9, 252. [Google Scholar] [CrossRef] [PubMed]
  165. Aval, S.F.; Lotfi, H.; Sheervalilou, R.; Zarghami, N. Tuning of Major Signaling Networks (TGF-β, Wnt, Notch and Hedgehog) by MiRNAs in Human Stem Cells Commitment to Different Lineages: Possible Clinical Application. Biomed. Pharmacother. 2017, 91, 849–860. [Google Scholar] [CrossRef]
  166. Shi, J.; Chi, S.; Xue, J.; Yang, J.; Li, F.; Liu, X. Emerging Role and Therapeutic Implication of Wnt Signaling Pathways in Autoimmune Diseases. J. Immunol. Res. 2016, 2016, 9392132. [Google Scholar] [CrossRef]
  167. Shang, X.; Böker, K.O.; Taheri, S.; Hawellek, T.; Lehmann, W.; Schilling, A.F. The Interaction between MicroRNAs and the Wnt/β-Catenin Signaling Pathway in Osteoarthritis. Int. J. Mol. Sci. 2021, 22, 9887. [Google Scholar] [CrossRef]
  168. Nadorp, B.; Soreq, H. Predicted Overlapping MicroRNA Regulators of Acetylcholine Packaging and Degradation in Neuroinflammation-Related Disorders. Front. Mol. Neurosci. 2014, 7, 9. [Google Scholar] [CrossRef] [PubMed]
  169. Komiya, Y.; Habas, R. Wnt Signal Transduction Pathways. Organogenesis 2008, 4, 68–75. [Google Scholar] [CrossRef] [PubMed]
  170. Marini, F.; Giusti, F.; Palmini, G.; Brandi, M.L. Role of Wnt Signaling and Sclerostin in Bone and as Therapeutic Targets in Skeletal Disorders. Osteoporos. Int. 2023, 34, 213–238. [Google Scholar] [CrossRef]
  171. Gao, Y.; Patil, S.; Jia, J. The Development of Molecular Biology of Osteoporosis. Int. J. Mol. Sci. 2021, 22, 8182. [Google Scholar] [CrossRef]
  172. Rudnicki, M.A.; Williams, B.O. Wnt Signaling in Bone and Muscle. Bone 2015, 80, 60–66. [Google Scholar] [CrossRef]
  173. Maes, M.; Kubera, M.; Kotańska, M. Aberrations in the Cross-Talks Among Redox, Nuclear Factor-κB, and Wnt/β-Catenin Pathway Signaling Underpin Myalgic Encephalomyelitis and Chronic Fatigue Syndrome. Front. Psychiatry 2022, 13, 822382. [Google Scholar] [CrossRef]
  174. D’Amico, R.; Cordaro, M.; Siracusa, R.; Impellizzeri, D.; Trovato Salinaro, A.; Scuto, M.; Ontario, M.L.; Crea, R.; Cuzzocrea, S.; Di Paola, R.; et al. Wnt/β-Catenin Pathway in Experimental Model of Fibromyalgia: Role of Hidrox®. Biomedicines 2021, 9, 1683. [Google Scholar] [CrossRef]
  175. Latremoliere, A.; Woolf, C.J. Central Sensitization: A Generator of Pain Hypersensitivity by Central Neural Plasticity. J. Pain 2009, 10, 895–926. [Google Scholar] [CrossRef]
  176. Zhao, Y.; Yang, Z. Effect of Wnt Signaling Pathway on Pathogenesis and Intervention of Neuropathic Pain. Exp. Ther. Med. 2018, 16, 3082–3088. [Google Scholar] [CrossRef] [PubMed]
  177. Martins, D.F.; Viseux, F.J.F.; Salm, D.C.; Ribeiro, A.C.A.; da Silva, H.K.L.; Seim, L.A.; Bittencourt, E.B.; Bianco, G.; Moré, A.O.O.; Reed, W.R.; et al. The Role of the Vagus Nerve in Fibromyalgia Syndrome. Neurosci. Biobehav. Rev. 2021, 131, 1136–1149. [Google Scholar] [CrossRef]
  178. Aster, H.-C.; Evdokimov, D.; Braun, A.; Üçeyler, N.; Sommer, C. Analgesic Medication in Fibromyalgia Patients: A Cross-Sectional Study. Pain Res. Manag. 2022, 2022, 1217717. [Google Scholar] [CrossRef] [PubMed]
  179. Spieker, J.; Frieß, J.L.; Sperling, L.; Thangaraj, G.; Vogel-Höpker, A.; Layer, P.G. Cholinergic Control of Bone Development and Beyond. Int. Immunopharmacol. 2020, 83, 106405. [Google Scholar] [CrossRef]
  180. Eimar, H.; Tamimi, I.; Murshed, M.; Tamimi, F. Cholinergic Regulation of Bone. J. Musculoskelet. Neuronal Interact. 2013, 13, 124–132. [Google Scholar] [PubMed]
  181. Ren, J.; Sang, Y.; Aballay, A. Cholinergic Receptor-Wnt Pathway Controls Immune Activation by Sensing Intestinal Dysfunction. Cell Rep. 2022, 41, 111575. [Google Scholar] [CrossRef] [PubMed]
  182. Labed, S.A.; Wani, K.A.; Jagadeesan, S.; Hakkim, A.; Najibi, M.; Irazoqui, J.E. Intestinal Epithelial Wnt Signaling Mediates Acetylcholine-Triggered Host Defense against Infection. Immunity 2018, 48, 963–978.e3. [Google Scholar] [CrossRef] [PubMed]
  183. Takahashi, T.; Shiraishi, A.; Murata, J. The Coordinated Activities of NAChR and Wnt Signaling Regulate Intestinal Stem Cell Function in Mice. Int. J. Mol. Sci. 2018, 19, 738. [Google Scholar] [CrossRef] [PubMed]
  184. Zendjabil, M.; Favard, S.; Tse, C.; Abbou, O.; Hainque, B. The microRNAs as biomarkers: What prospects? C. R. Biol. 2017, 340, 114–131. [Google Scholar] [CrossRef] [PubMed]
  185. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef]
Figure 1. PRISMA Extension for Scoping Reviews (PRISMA-ScR) flowchart. a: comment, meta-analysis, review, meeting abstract; N = number of articles.
Figure 1. PRISMA Extension for Scoping Reviews (PRISMA-ScR) flowchart. a: comment, meta-analysis, review, meeting abstract; N = number of articles.
Ijms 24 13513 g001
Figure 2. Summary diagram of dysregulated miRNAs in both osteoporosis (OP) and fibromyalgia (FM). N = number of articles; n = number of miRNAs.
Figure 2. Summary diagram of dysregulated miRNAs in both osteoporosis (OP) and fibromyalgia (FM). N = number of articles; n = number of miRNAs.
Ijms 24 13513 g002
Figure 3. Summary of miRNAs found in fibromyalgia (orange circle) and in osteoporosis (blue circle). “↑” miRNAs at the upper part of the circles are upregulated. “↓” miRNAs at the lower part of the circles are downregulated. “↑↓” miRNAs at the equator of the circles are either up- or downregulated. Intersection of circles in green represents miRNAs common for FM and OP. References are listed in Tables S1 and S2.
Figure 3. Summary of miRNAs found in fibromyalgia (orange circle) and in osteoporosis (blue circle). “↑” miRNAs at the upper part of the circles are upregulated. “↓” miRNAs at the lower part of the circles are downregulated. “↑↓” miRNAs at the equator of the circles are either up- or downregulated. Intersection of circles in green represents miRNAs common for FM and OP. References are listed in Tables S1 and S2.
Ijms 24 13513 g003
Figure 4. Common miRNAs in fibromyalgia and in osteoporosis and regulation via the Wnt signaling pathway and/or the cholinergic system. References are listed in Table 1 and Table 2. (↓ = downregulated; ↑ = upregulated).
Figure 4. Common miRNAs in fibromyalgia and in osteoporosis and regulation via the Wnt signaling pathway and/or the cholinergic system. References are listed in Table 1 and Table 2. (↓ = downregulated; ↑ = upregulated).
Ijms 24 13513 g004
Table 1. Differentially expressed miRNAs in fibromyalgia (FM) and osteoporosis (OP). (↓ = downregulated; ↑ = upregulated).
Table 1. Differentially expressed miRNAs in fibromyalgia (FM) and osteoporosis (OP). (↓ = downregulated; ↑ = upregulated).
miRNAs Expressed in Same Direction in FM and OP (n = 7).miRNAs Expressed in Opposite Direction in FM and OP (n = 9)
miRNAsmiRNA
Expression
ReferencesmiRNAsmiRNA
Expression
References
miR-1-(3p)FM[28]miR-7-5pFM[30]
OP[36,37]OP[42]
let-7a-(3p or 5p)FM[29]miR-23b-(3p)FM[31]
OP[37]OP[44,45]
miR-9-(3p or 5p)FM[30]miR-107FM[29,33]
OP[38,39]OP[46]
miR-29a-3pFM[31]miR-139-5pFM[28]
OP[22]OP[44]
miR-128-(3p)FM[30]miR-143-(3p)FM[34]
OP[40]OP[41,47]
miR-328-3pFM[30]miR-151a-(3p or 5p)FM[29]
OP[41]OP[48,49]
miR-335-5pFM[32]miR-186-5pFM[30]
OP[42,43]OP[42]
miR-338-3pFM[34]
OP[50]
miR-766-(3p or 5p)FM[30]
OP[51]
Table 2. Characteristics of the 7 common miRNAs in fibromyalgia (FM) and osteoporosis (OP).
Table 2. Characteristics of the 7 common miRNAs in fibromyalgia (FM) and osteoporosis (OP).
miRsPathologyReferenceFindings
miR-1(-3p)FM[28]Myoblast differentiation - Downregulation of miR-1(-3p) following reduced physical activity in FM patients because of pain and fatigue symptoms.
Modulation of brain-derived neurotrophic factor (BDNF) expression. BDNF in skeletal muscle inhibits myogenic differentiation.
OP[36]Balance between osteogenesis and adipogenesis of mesenchymal stem cells (MSC). Upregulation of miR-1-3p during osteogenesis and downregulation during adipogenesis. Secreted Frizzled-related protein 1 (SFRP1) is a direct target of miR-1-3p. Inhibition of miR-1-3p decreased bone formation and bone mass.
Regulation of myostatin gene. Influence on muscle hypertrophy.
Reduced/suppressed expression of fibronectin 1, BDNF, Dickkopf 1 (DKK1). Suppressed proliferation and migration of oral squamous cell carcinoma/ renal cell lines.
[37]Regulation of RAB5C expression. hsa-miR-1-3p and his target mRNA RAB5C may play a critical role in the bone metabolism of postmenopausal osteoporosis.
Regulation in skeletal tissue.Muscle proliferation, muscle differentiation and myogenesis.
let-7a(-3p or 5p)FM[29]Repression of μ-opioid receptor expression.Regulation of the endogenous opioid system and in opioid tolerance.
OP[37]Downregulated.
miR-9(-3p or 5 p)FM[30]CholinomiRNA. Regulation of Janus kinase 2 (JAK2) expression. JAK2 is pivotal for IL6/JAK2/STAT3 axis-mediated inflammation.CholinomiRs may shift inflammatory processes via modulation of the systemic cholinergic system.
OP[38]Inhibition of osteogenic differentiation of hMSCs.Inhibiting the miR-9-5p expression promoted the expression of osteocalcin, runt-related transcription factor 2 (Runx2) and bone morphogenetic protein 7 (BMP-7), enhanced BMD, and promoted fracture healing.
Inhibition of skeletal cell proliferation and differentiation.
[39]Inhibition of the expressions of osteogenic-related genes.Direct binding to Wnt3a. Wnt3a overexpression partially reversed the regulatory effect on osteogenic differentiation of MSCs.
Promotion of adipogenic-related genes expression.
miR-29a-3pFM[31]Upregulation during aging in mice and decreased in several pathologies including muscular dystrophy type 1.
OP[23]Induction of beta-catenin protein levels.Activation of canonical Wnt signaling.
Key regulation of collagen expression.
miR-128-3pFM[30]CholinomiRNA- Upregulation and Involvement in the neuronal oxidative stress response.Modulation of the cholinergic system.
OP[40]Inhibition of osteoblast differentiation.Down-regulation of sirtuine 6 (SIRT6) expression.
miR-328-3pFM[30]CholinomiRNA.Modulation of the cholinergic system.
OP[41]Inhibition of osteoblast differentiation.
miR-335-5pFM[32]Sole miRNA that differed significantly from controls.
OP[42]Upregulation in osteoporosis with low-traumatic fractures compared to controls.
[43]Upregulation in osteoporosis with vertebral fractures/low BMD compared to low BMD/no fractures and controls.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Philippe, S.; Delay, M.; Macian, N.; Morel, V.; Pickering, M.-E. Common miRNAs of Osteoporosis and Fibromyalgia: A Review. Int. J. Mol. Sci. 2023, 24, 13513. https://doi.org/10.3390/ijms241713513

AMA Style

Philippe S, Delay M, Macian N, Morel V, Pickering M-E. Common miRNAs of Osteoporosis and Fibromyalgia: A Review. International Journal of Molecular Sciences. 2023; 24(17):13513. https://doi.org/10.3390/ijms241713513

Chicago/Turabian Style

Philippe, Soline, Marine Delay, Nicolas Macian, Véronique Morel, and Marie-Eva Pickering. 2023. "Common miRNAs of Osteoporosis and Fibromyalgia: A Review" International Journal of Molecular Sciences 24, no. 17: 13513. https://doi.org/10.3390/ijms241713513

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