Skip to main content

The role of miRNA and lncRNA in heterotopic ossification pathogenesis

Abstract

Heterotopic ossification (HO) is the formation of bone in non-osseous tissues, such as skeletal muscles. The HO could have a genetic or a non-genetic (acquired) background, that is, it could be caused by musculoskeletal trauma, such as burns, fractures, joint arthroplasty (traumatic HO), or cerebral or spinal insult (neurogenetic HO). HO formation is caused by the differentiation of stem or progenitor cells induced by local or systemic imbalances. The main factors described so far in HO induction are TGFβ1, BMPs, activin A, oncostatin M, substance P, neurotrophin-3, and WNT. In addition, dysregulation of noncoding RNAs, such as microRNA or long noncoding RNA, homeostasis may play an important role in the development of HO. For example, decreased expression of miRNA-630, which is responsible for the endothelial–mesenchymal transition, was observed in HO patients. The reduced level of miRNA-421 in patients with humeral fracture was shown to be associated with overexpression of BMP2 and a higher rate of HO occurrence. Down-regulation of miRNA-203 increased the expression of runt-related transcription factor 2 (RUNX2), a crucial regulator of osteoblast differentiation. Thus, understanding the various functions of noncoding RNAs can reveal potential targets for the prevention or treatment of HO.

Introduction

Heterotopic ossification: a brief overview

Heterotopic ossification (HO) is a dysregulation of skeletal muscle homeostasis and regeneration that leads to the formation of mature bone in unusual locations. HO develops in skeletal muscles and surrounding tissues, such as fascia, tendons, skin, and subcutis. Most lesions are small and clinically irrelevant, but extensive HO can limit patient physical functioning and quality of life [1]. The clinical manifestations vary depending on the stage of HO development. Typical clinical symptoms in the early phases are such as localized pain, tenderness, and swelling. At later stages, a limited range of motion (ROM) may affect the joint, resulting in complete ankylosis in the most severe cases [2].

Acquired heterotopic ossification

HO formation may occur due to fractures, extensive soft tissue damage, burns, amputations, and combat-related injuries. It can also be triggered by iatrogenic trauma associated with the surgical approach. Up to 90% of acetabulum fractures and 8.6% of distal humerus fractures subjected to surgical treatment result in the formation of HO [3]. It can also occur after arthroscopic procedures and after hip arthroscopy was detected in up to 46% of cases [4]. Approximately 29.9% of patients with total hip arthroplasty (THA) developed HO, but most cases were asymptomatic. However, large lesions occurring in 0.57–2.7% of patients can significantly limit surgery benefits [5]. Extensive HO can influence patient life quality assessed by patient-reported outcome measures (PROMs). It is worth mentioning that mature HO rarely causes pain. However, they substantially reduce the ROM in the affected joint [6]. Patients who were subjected to total hip arthroplasty (THA) with extensive HO lesions (Brooker III, IV) do not benefit from surgery in the ROM aspect, as compared to the preoperative status [7]. HO has been reported to significantly limit joint ROM after revision knee arthroplasty [8], burns [9], and elbow fractures [10]. However, even severe ROM-limiting HOs usually do not cause pain [11].

Another example of acquired HO is neurogenic heterotopic ossification (NHO). The NHO may form around the hip, knee, elbow, and glenohumeral joint due to the response to neuroinflammation signals and systemic changes caused by central nervous system (CNS) injury. Joint mobility problems resulting from NHO often cause nursing challenges [12]. The presence of NHO is associated with a poor functional outcome in patients after CNS injury [13]. In such cases, restricted ROM is the major problem, as pain is often absent due to sensory deficits [14]

Recently, HO has been reported in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection who require mechanical ventilation. The mechanism of HO in COVID-19 patients is unclear. However, prolonged immobilization, global inflammation, and cytokine storm are likely to be the triggering factors [15, 16].

Zhang et al. described another subtype of HO—heterotopic ossification of the tendon and ligament (HOTL). HOTL includes ossification of the posterior longitudinal ligament of the spine (OPLL) and calcific tendinitis [17]. However, usually OPLL and calcific tendinitis are not mentioned in reviews focusing on HO [1, 18, 19] but are described separately [20, 21]. OPLL can result in radiculopathy or myelopathy that causes spasticity and gait disturbances [17]. It is the most common in the cervical spine. OPLL affects approximately 0.1–4.3% of the world population, with a high prevalence in Asian populations [22]. Unlike other types of acquired HO, it does not result from trauma. The risk factors for OPLL are both genetic, e.g., COL11A2 [23] and COL6A1 polymorphisms [24], and environmental, i.e., high-sodium diet [25], obesity [26], and nonalcoholic fatty liver disease [27]. The role of ncRNAs in OPLL is broadly described in a review by Yuan et al. [28]. The calcific tendinitis occurs when repetitive microtrauma acts on the tendons, e.g., in athletes or manual workers. It is common in the rotator cuff tendons [29]. The lesions resemble incomplete ossification and do not contain mature bone but amorphous calcium deposits [30], so they should not be considered a HO subtype.

Genetic heterotopic ossification

Heterotopic ossification may be the most notable clinical characteristic of three genetic diseases: fibrodysplasia ossificans progressiva (FOP), progressive osseous heteroplasia (POH), and hereditary Albright osteodystrophy (AHO). Genetic HOs are very severe but rare conditions and belong to the so-called orphan disease family. Osteogenesis induction in FOP is caused by a mutation of the activin A receptor type 1/activin receptor-like kinase 2 gene (ACVR1/ALK2; in most cases R206H), which encodes the bone morphogenetic protein (BMP) receptor, type I. POH and AHO are caused by mutations in the GNAS1 gene, influencing Wingless-related integration site (WNT) and Hedgehog signaling (HH), the key controllers of skeletal maturation and regeneration [31,32,33]. The prevalence of FOP ranges from 0.4 to 65 cases per 10,000 [34] and the prevalence of AHO is 7.2 cases per million [35]. The epidemiology of POH remains unknown, as less than 60 cases have been reported, so far [36]. Among ossifications of genetic origin, the most severe are those occurring in patients suffering from FOP. FOP is characterized by periodic exacerbations with localized painful soft tissue inflammation that leads to the development of HO in muscles, joints, tendons, and ligaments. Over time, it leads to severe joint limitations and loss of ROM. Most FOP patients are wheelchair-bound in the third decade of life [37]; in addition, they report emotional problems, such as anxiety, depression, or irritability. The severity of pain associated with FOP significantly influences emotional health and overall quality of life [38].

Possible mechanism of heterotopic ossification

Although many research projects have been devoted to the description of HO, including histological descriptions of the lesions, their progression, and risk factors, the exact molecular processes remain unknown [39]. Patients with HO are most likely to have global changes at the multi-omics level [40], including genetic [41,42,43], epigenetic [44], transcriptomic, proteomic [45, 46], and metabolic processes [47]. In particular, both genetic HO and acquired HO have already been represented in animal models. The existence of an animal model that satisfies human disease conditions is essential for a detailed explanation of the molecular and cellular mechanisms responsible for disease progression and the preclinical evaluation of potential promising therapeutic tools [31, 39].

Many lines of evidence indicate that the development of HO in skeletal muscle may be the result of pathological differentiation of stem or progenitor cells present in skeletal muscle [39]. However, the identity of these cells is not yet clear. Animal studies suggest that progenitor cells responsible for pathological osteogenesis may differ depending on the HO subtype. Research involving mouse models indicates that endothelial cells, mesenchymal cells, pericytes, tendon, and other connective tissue cells, or circulating stem cells may be the source of HO precursors [48, 49]. In both acquired and genetic HO, the differentiation of precursor cells is initiated by inflammatory cells, including lymphocytes, macrophages, and mast cells [50, 51]. It is accompanied by the release of cytokines and growth factors by immune cells including interleukin 1β (IL-1β), interleukin 6 (IL-6), oncostatin M (OSM), neurotrophin-3 (NT-3), activin A, BMP, transforming growth factor β (TGFβ), and substance P (SP) [50,51,52]. Differentiation of precursor cells leading to the formation of HO is complemented by increased translation of proteomic biomarkers of HO, such as alkaline phosphatase (ALP), osteocalcin (OCN), osteopontin (OPN/SSP1), and bone sialoprotein (BSP) [45].

The HO can form as any other bone through endochondral or intramembranous ossification. Both processes occur during mammalian skeletal development and bone remodeling during fracture healing [53]. Endochondral ossification occurs in cartilage models of long bones when hypertrophic chondrocytes produce an extracellular matrix that is mineralized in the ossification centers [54]. HO develops through endochondral osteogenesis, which is preceded by infiltration and migration of lymphocytes, fibroproliferation, neovascularity, and cartilage formation [55]. Similarly, in FOP the HO develops with endochondral ossification. Impaired osteochondrogenesis in FOP results not only in extraskeletal bone formation, but also in growth plate dysplasia, early osteoarthritis, and joint deformation [56]. The flare-ups in FOP are accompanied by an elevation of serum cartilage-derived retinoic acid-sensitive protein (CD-RAP), which is a biomarker of chondrogenesis [57]. Opposing to FOP [58] POH ossifications are formed by an intramembranous process in which mesenchymal cells differentiate directly into osteoblasts and form ossification centers [59]. During embryogenesis Intramembranous ossification contributes to the development of the skull, mandible, and middle part of the clavicle [60], and is also responsible for pathological bony bridge formation in growth plate injuries [61].

The precise transduction of signals in HO remains unclear. One of the best-understood regulators of bone development is BMPs, factors from the TGFβ superfamily. BMPs are ligands of transmembrane BMP receptor type I (e.g., ACVR1/ALK2) and type II. From BMP, the signal is transduced with SMAD or non-SMAD pathways. SMADs are cytoplasmic proteins activated by phosphorylation that transmit the signal to the nucleus. The non-SMAD-dependent pathway involves the activation of mitogen-activated protein kinase (MAPK). The signal transmitted by both SMAD and non-SMAD pathways leads to the expression of osteogenesis- and chondrogenesis-promoting transcription factors, such as runt-related transcription factor 2 (RUNX2), Osterix (OSX) or distal-less homeobox 5 (DLX5) [39].

Another mechanism associated with the development of HO that could explain the formation of traumatic HO includes the regulation of the immune response by changes in nuclear factor-κB (NF-κB) expression. The NF-κB induces osteogenesis in response to pro-inflammatory ligands of the toll-like receptor (TLR). TLR recognizes damage-associated molecular patterns (DAMPs) released from cells or extracellular matrix after injury, for example, heat shock protein (HSP), high-mobility group box 1 (HMGB1), hyaluronan, or it can be activated by pathogen-associated molecular patterns (PAMPs), for example, lipopolysaccharide (LPS) [62]. Other studies investigated the role of the hypoxic microenvironment in the HO development. Overexpression of hypoxia-inducible factor 1α (HIF-1α), a key transcriptional controller of the hypoxic cellular response, may play an important role in pathological bone formation after tissue damage [63]. The rapamycin mammalian target (mTOR) signaling pathway was recently identified to play a role in the pathological osteogenesis [64]. The mTOR is a nutrient sensor and a controller of protein synthesis. The mTOR protein complex-1 (mTORC1) influences cell growth, survival, and proliferation in response to oxygen level, energy status, growth factors, amino acids level [65] or mechanical stimulation [2]. Other factors such as HH and WNT/β-catenin pathways responsible for skeletal maturation and regeneration are also affected by genetic and acquired HO [66]. There is evidence that these pathways can cross talk with BMP signaling, but this network is still being investigated [39, 64, 66].

Recent studies suggest that the development of HO is also regulated by noncoding RNAs. By changing gene expression and mRNA degradation, noncoding RNAs can indirectly stimulate or inhibit the HO formation [45, 67]. Understanding the role of noncoding RNA in HO could lead to highly targeted and efficient HO therapies based on molecules from this group [68].

Current concepts in HO treatment

Today, pharmacological anti-inflammatory treatment and radiation therapy or both are used to prevent HO. Non-steroidal anti-inflammatory drugs (NSAIDs) are the most frequently administered pharmacological prophylaxis of acquired HO. Recent meta-analysis by Migliorini et al. supports the use of celecoxib, diclofenac, or naproxen in the prevention of HO due to their high effectiveness compared to other NSAIDs [69]. The efficiency of such drugs, both non-selective and selective cyclooxygenase (COX) inhibitors, has been proven in HO prevention. However, their action is not specifically targeted against HO, but rather by inhibiting arachidonic acid prostaglandin production, NSAIDs cause suppression of the inflammatory mechanisms involved in HO [70]. NSAIDs also suppress the transcription factor NF-κB—an immune response regulator that can control osteogenesis [71].

Radiotherapy (RT) is proven to be effective in hip joint surgery [72], non-hip sites such as the elbow or knee [73], and NHO prophylaxis after spinal cord injury [74]. RT is administered before surgery or within 72 h after surgery [75]. According to a meta-analysis by Milakovic et al., there is no difference between postoperative or preoperative RT in preventing HO progression. RT doses higher than 2500 cGy do not result in a better outcome [76]. A single RT dose is less effective than divided to multiple fractions in the prevention of HO after THA [77]. The potential side effects include fatigue, wound healing delay, swollen joints, and very rarely neoplasms that are secondary to RT [78]. Several cases of radiation-induced sarcoma have been reported so far after HO prophylaxis [79,80,81]. Radiation therapy does not appear to influence implant loosening after THA [82]. There is no consensus, however, as to whether NSAIDs are more effective than RT in HO prevention. A meta-analysis by Pakos et al., focusing on seven randomized trials involving a total of 1143 patients after major hip procedures (THA, acetabular fracture surgery), documented that RT was nearly twice as effective as NSAIDs in HO prophylaxis (Brooker III, IV) [72]. Another meta-analysis by Shapira et al. presented opposing results [83]. Undoubtedly, the cost-effectiveness of HO prophylaxis favors NSAID over RT [84]. The theory that could explain the efficacy of RT in HO prevention is the inhibition of osteogenic differentiation of the MSC. It is accompanied by a decrease in RUNX2 expression and a decrease in ALP and OCN levels [85].

Experimental approaches directly targeting molecular and signaling processes have been tested in vivo, in animal models, and in humans [86]. Human studies focus on the severe form of HO to FOP. Currently, the most promising effects of these studies in HO inhibition are those regarding the BMP pathway, including receptor ACVR1/ALK2. Another approach is to target NF-κB or mTOR signaling [70].

The clinical trial (LUMINA-1) investigated antibodies against activin A in adult patients with FOP. Garetosmab is a human antibody that binds to activin A, an agonistic ligand of ACVR1/ALK2 [87]. The initial results of the phase 2 study indicate that Garetosmab can reduce the formation of new HO. However, drug administration was halted due to serious fetal adverse events [88, 89]. Recently, a phase 3 trial (OPTIMA) of Garetosmab was registered for adults with FOP [90]. Another monoclonal antibody (DS-6016a) targeted against ACVR1/ALK2 is tested in a phase 1 study in healthy volunteers [91]. Recently orally administered small molecule inhibitors of ACVR1/ALK2 [92] were tested in phase 1 studies on healthy individuals (KER-047 [93], BCX9250 [94]) and phase 2 studies in FOP patients (IPN60130 (FALKON) [95], INCB000928 (PROGRESS) [96]). Dorsomorphin and LDN-193189 are other molecules that inhibit ACVR1/ALK2 but their action is not specific, thus, the potential safety profile of these drugs is questionable [97, 98]. Recent studies in mice have indicated that systemic administration of a neutralizing antibody to activin A inhibits acquired forms of HO also, expanding the therapeutic repertoire of this immunological treatment [99].

Another option for HO drug research is to investigate treatments whose safety profile has already been assessed and which were successful in the therapy of other diseases rather than in new drug development [89]. Anti-leukemic Saracatinib is currently being investigated in adults with FOP (phase 2 trial (STOPFOP)) [100]. It is a potent inhibitor that binds to the ATP pocket of the ACVR1/ALK2 kinase domain and also blocks SMAD phosphorylation and transduction of osteochondrogenic signaling [101, 102]. Another drug repurposed in HO is rapamycin (Sirolimus) which is used to prevent transplant rejection. It influences mTOR signaling that modulates ACVR1/ALK2 and HIF1α action during chondrogenesis in HO [103], effectively blocking HO development in the FOP mouse model [104]. It is currently being investigated in phase 2 in adult FOP patients [105]. Palovarotene, a retinoic acid receptor gamma (RARγ) agonist, previously used in patients with emphysema [106], can inhibit the chondrogenesis phase in HO formation. Palovarotene influences BMP/SMAD-dependent pathway [107] and NF-κB signaling [108] preventing HO formation via endochondral ossification. In a trauma-induced HO rat model. Palovarotene inhibited HO formation and caused down-regulation of chondrogenesis biomarkers, i.e., SRY-box transcription factor 9 (SOX9) and osteogenesis biomarkers (OCN, RUNX2) [109]. It was already tested in adult FOP patients, and a phase 3 clinical trial phase 3 is ongoing (MOVE) [110]. The ongoing clinical trials of FOP drugs are summarized in Table 1.

Table 1 Ongoing clinical trials in fibrodysplasia ossificans progressiva

Most studies focus on the prevention of HO, and when mature lesions have already formed, the only treatment method is surgical excision [70]. An innovative approach uses osteoclasts, cells responsible for bone remodeling, against mature HO. Osteoclasts were modified with tetracycline, which has a high affinity for bone hydroxyapatite. Artificially engineered osteoclasts with a high affinity for calcified bone capable of HO resorption were successful in treating already formed lesions in tenotomy, intramuscular, and genetic HO mouse models [111].

In recent years, great progress has been made in genetic HO treatment [32, 88, 101]. Recently (January 2022), Health Canada approved palovarotene (Sohonos) as the first drug for Fibrodysplasia Ossificans Progressiva (FOP) [112]. For acquired HO, NSAIDs and RT are still the methods of choice [69]. Emerging evidence indicates the importance of noncoding RNA in the pathogenesis of HO. It suggests that a clinically relevant noncoding RNA signature may be detected in patients with certain risk factors and could be used to predict or prevent HO [113].

Advances in ncRNA therapy

Noncoding RNAs (ncRNAs) do not encode proteins but play an important role in many biological processes, such as the regulation of gene expression, RNA processing, or protein synthesis. The group of ncRNAs includes, for example, ribosomal RNAs (rRNAs) and transfer RNAs (tRNAs) involved in protein translation, small nucleolar RNAs (snoRNAs) regulating rRNA biogenesis, and small nuclear RNAs (snRNAs) participating in mRNA splicing (broadly reviewed in [117, 118]). Recently, the role of ncRNAs, such as small interference RNA (siRNA), has been intensively investigated in the context of diseases related to the musculoskeletal system [119]. Moreover, ncRNAs have the potential to be applied in the therapy [120]. Here, we focus on two groups of ncRNAs, i.e., microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), which were described to be involved in the formation and progression of HO.

microRNA (miRNA)

MicroRNAs are short RNA molecules that consist of approximately 18–30 nucleotides and play an important role in posttranscriptional RNA silencing. Mature miRNA binds to the 3’untranslated region (3’UTR) of its target mRNA, which leads to destabilization or degradation of the mRNA. Regardless of the mechanism of action, the effect of miRNA activity is to prevent protein synthesis coded by target mRNA. Although most of the data underline the inhibitory properties of miRNAs, it should be mentioned that some studies showed that miRNA activity can also lead to up-regulation of gene expression [121,122,123,124,125,126,127]. This activation involves argonaute RISC catalytic component 2 gene (AGO2) and fragile X mental retardation-related protein 1 gene (FXR1) instead of GW182 and was observed in quiescent (G0) somatic cells and frog oocytes [128, 129]. miR-10a can serve as an example of miRNA-dependent up-regulation of gene expression. Its binding to 5’UTR increased the translation of mRNAs encoding ribosomal proteins during amino acid starvation of mouse embryonic stem cells [130].

The biogenesis of miRNA is quite complex and involves many cellular mechanisms (Fig. 1.). The primary miRNA transcript (pri-miRNA) is a long molecule with a characteristic local hairpin structure within which a mature miRNA sequence is present. Pri-miRNA is cleaved by the endonuclease DROSHA, which, together with the RNA binding protein Di George syndrome critical-related gene 8 (DGCR8), form the microprocessor complex [131,132,133,134]. Cropping of the pri-miRNA leads to the formation of pre-miRNA. However, not all miRNA biogenesis involves the Microprocessor complex. Some miRNAs are encoded in so-called mirtrons present in pre-mRNA introns [135,136,137,138]. Thus, these pre-miRNAs are generated during pre-mRNA splicing. Furthermore, pre-miRNAs are exported from the nucleus to the cytoplasm by exportin 5 (XPO5)/RanGTP complex [139,140,141]. In the cytoplasm, pre-miRNA is further processed by other endonuclease DICER, which results in the formation of a miRNA duplex [142,143,144]. Although both miRNA strands can be functional, during biogenesis one of them is degraded and the other forms an RNA-induced silencing complex (RISC) together with AGO and GW182 proteins [145,146,147,148,149]. After forming the RISC complex, some specific regions of the miRNA structure can be distinguished. Among them, the most important is the “seed” domain, which is directly responsible for recognizing target mRNA. Binding of RISC to mRNA results in mRNA destabilization, mRNA degradation, or inhibition of translation [150]. Furthermore, multiple studies have reported that miRNAs not only act within the cell cytoplasm but also are released into extracellular fluids. These extracellular miRNAs have the potential to be used as biomarkers for diverse conditions [151,152,153]. Extracellular miRNAs can be delivered to target cells and have the potential to act as autocrine, paracrine, and/or endocrine regulators that modulate cellular activity.

Fig. 1
figure 1

miRNA biogenesis and mechanism of action. miRNA is transcribed by POL II, leading to the generation of often polycistronic pri-miRNA. Pri-miRNA is cleaved by the microprocessor complex which consists of DROSHA and DGCR8 and results in the release of small hairpin-shaped RNA, called pre-miRNA. The pre-miRNA is exported to the cytoplasm by the XPO5/RanGTP complex and cleaved by DICER. DICER cleavage results in the release of a miRNA duplex. One of the miRNA strands becomes degraded, and the other forms a RISC complex with proteins from the AGO and GW182 families. Mature miRNA acts in RISC, and its activity leads to mRNA degradation, mRNA destabilization by deadenylation and decapping, or inhibition of translation. The figure was created for this article; it is not based on any previously published image

Long noncoding RNA (lncRNA)

Long noncoding RNAs are a very heterogeneous group of ncRNAs, generated via pathways similar to mRNAs and characterized by a minimum size of 200 nucleotides (Fig. 2). In fact, the length of the molecule is the only common feature of all lncRNAs. These transcripts play diverse roles within the cell, acting in both the nucleus and the cytoplasm. However, three main mechanisms of lncRNA activity can be distinguished [117, 118]. In the nucleus, lncRNAs were shown to be involved in the regulation of chromosome structure and mediating chromatin remodeling by recruiting histone-modifying complexes such as histone acetylases or deacetylases (HDACs) (e.g., [154,155,156]; reviewed in [157]). Furthermore, lncRNAs can directly affect gene expression by binding to enhancer sites or transcription factors [158,159,160]. lncRNAs acting in the cytoplasm are responsible for posttranscriptional regulation of gene expression by modulating the accessibility or stability of mRNA [117, 118]. Finally, cytoplasmic lncRNAs can directly bind to and inhibit miRNA-dependent gene silencing by functioning as a competing RNA [161, 162]. Therefore, lncRNAs appear to be important regulators of many processes, from the organization of chromosome and chromatin remodeling to the posttranscriptional regulation of gene expression in the cytoplasm.

Fig. 2
figure 2

Mechanisms of lncRNA action. Nuclear lncRNAs regulate gene expression by: A regulating chromatin remodeling; B recruiting chromatin-modifying complexes (such as HDACs); C regulating transcription factor activity. Furthermore, cytoplasmic lncRNAs impact mRNA processing through: D regulating mRNA stability; E regulating translation; F acting as competitors for miRNAs; and G protein function by binding proteins or peptides. The figure was created for this article; it is not based on any previously published image

miRNAs and lncRNAs as potential therapeutics

Noncoding RNAs represent very promising tools for the treatment of various diseases [163]. Eleven RNA-based therapeutics have already been approved by the FDA and/or the European Medicines Agency (EMA) [163]. These therapeutics are small interfering RNAs (siRNAs) or antisense oligonucleotides (ASO). They are used, for example, in the therapy of spinal muscular atrophy, Duchene muscular dystrophy, or homozygous familial hypercholesterolemia [163]. Moreover, numerous RNAs are clinically tested. Some of the RNA-based therapies (phase 2 or 3 clinical trials) involve miRNA. However, no lncRNA-based therapy has been available to the clinic, so far [163]. The function of miRNA-based therapeutics relies on restoring or depleting the miRNA or inhibiting their interactions with targets. Few types of miRNA-based molecules are currently being investigated as therapeutics. First, miRNA mimics that have the same sequence as endogenous miRNA and mimic their function. Currently, one molecule, i.e., miR29 mimic, is tested in clinical trials for the treatment of pathological skin fibrosis (NCT02603224, nct03601052). The second type of molecules are antagomiRs that are antisense to specific miRNAs and prevent their interactions with targets. Two such molecules are currently being investigated in clinical trials, these are anti-miR103/107 (NCT020612662 and NCT02826525) and anti-miR122 (NCT01646489, NCT01727934, NCT01872936, and NCT01200420). They are tested in the treatment of type 2 diabetes and hepatitis C virus infection, respectively. Many other miRNA- or lncRNA-based therapeutics are also studied, for example, in the treatment of resistance to cancer therapy [164] and other diseases [165].

miRNAs and lncRNAs as potential biomarkers

Several HO biomarkers have been proposed to date, but there is no consensus which of them should be used in clinical practice [166]. Physiological bone turnover indicators based on protein levels are not specific and can change due to other health conditions [167]. Thus, ncRNAs are a promising new group of biomarkers that can be easily isolated from all body fluids and were suggested to be specific markers in many other diseases [168,169,170,171,172]. However, before ncRNA could be widely used as a diagnostic tool, the problem of low expression levels, instability [172], and sequencing costs [208] had to be overcome. Once the mechanism of the complex ncRNA interactions will be uncovered, validation studies and the establishment of cutoff values are needed to enable the application of ncRNA as reliable HO biomarkers. In addition, their specificity as biomarkers should also be investigated in other diseases and reproducible measurement methods should be implemented.

The role of miRNA and lncRNA in heterotopic ossification

Dysregulation of the miRNA expression profile in HO

Recently, the role of miRNA and lncRNA has been intensively investigated to understand the background of diseases related to the musculoskeletal system [119] as well as to identify novel drug targets [120]. However, the knowledge about the role of these molecules in the formation of HO is very limited. The role of some of them has been described in the formation of HO resulting from mutations or different types of injury. The study by Ji et al., in which the level of miRNA was compared between patients asymptomatic for HO and those who developed HO, showed that miR205 and miRNA-215 were upregulated and muscle-specific miRNAs, that is, miR1, miR26a, miR133a, miR133b, miR146b, and miR206 were dysregulated [173]. miR205 was described mainly in myoepithelial cells and many cancer types [174] while miR215 was found in osteosarcoma [175]. The other ones which had expression changes were described in this study, i.e., miR1, miR26a, miR133a, miR133b, miR146b, and miR206 belong to muscle-specific miRNAs known as myomiRs. They play an important role in the regeneration of skeletal muscle, including activation, proliferation, and differentiation of muscle stem cells, i.e., satellite cells [176]. Thus, disrupted expression of myomiRs during HO development may be related to defective differentiation of myogenic cells [176].

Comparison of the miRNA profile in the serum of patients with immature HO and mature HO allowed the identification of miR630 as a factor that may be involved in the development of these pathologies [177]. The level of miR630 was significantly lower in the case of both types of HO. Thus, it was suggested that this molecule could serve as an early marker of HO formation [177]. Interestingly, miR630 and its direct target Slug, that is, a member of the Snail family of zinc finger transcription factors, was involved in the endothelial–mesenchymal transition of endothelial cells, which were suggested to be a source of cells responsible for HO formation [177]. Down-regulation of miR630 simultaneously with BMP4 and TGFβ2 treatment increased endothelial cells’ osteogenic differentiation [177].

Another molecule, identified by comparison of miRNAs between the normal bone in patients and primary and mature HO, was miR203 [68]. The level of miR203 decreased in HO; simultaneously, the level of RUNX2 was increased. miR203 directly targets RUNX2 [68]. Furthermore, up-regulation of miR203 inhibited osteogenic differentiation of human osteoblasts. Interestingly, a chemically modified miRNA mimic, named agomiR203, injected into mice that underwent a tenotomy to generate a traumatic HO model, significantly decreased the development of HO, compared to control mice, i.e., injected with phosphate-buffered saline [68].

Next, miR421 is associated with cell proliferation and cancer [178, 179]. However, patients with HO show significantly lower expression of miR421 in their bone and blood, compared to patients who did not develop HO. Thus, miR421 could play a regulatory role in the HO induction [180]. It was documented that BMP2, i.e., the strongest osteogenic induction factor, was the direct target of miR421. Verification of the role of BMP2 in patients with HO induced by humeral fracture showed a significantly increased expression of BMP2 in ossified tissues and blood. Therefore, the development of HO may be related to the up-regulation of BMP2 and down-regulation of miR421 [180].

On the other hand, miR433 is related to numerous diseases. Its expression is increased in fibrotic heart disease rat models [181]. Furthermore, miR433 by targeting the RAP1a and MAPK signaling pathway, miR433 inhibits cancer cell proliferation [182]. Additionally, miR433 plays an important role in esophageal cancer and glioma [183, 184]. Bioinformatics prediction showed that miR433 is a potential upstream regulator of OPN/SPP1 [185]. OPN/SPP1 is a pro-inflammatory factor that affects the adhesion and proliferation of synovial cells [186]. OPN/SPP1 is overexpressed in the cartilage and synovium of osteoarthritis patients with osteoarthritis [187, 188]. The expression of OPN/SPP1 is regulated, among others, by microRNAs, including miR433. This molecule affects the expression of OPN/SPP1 by direct binding to the 3′‑UTR of OPN/SPP1 mRNA [189]. In the case of patients with callus and HO in patients with traumatic brain injury (TBI), the expression of OPN/SPP1 increased significantly and miR433 was reduced, leading to increased accumulation of pro-inflammatory OPN/SPP1 protein in bone tissues and negatively affecting tibial fracture healing of tibial fractures [189].

De Vasconcellos et al. analyzed the miRNA expression profile in samples of patients developing HO and healthy individuals. HO patients demonstrated a unique molecular signature, that is, significantly upregulated miR1, miR26a, miR125b, miR133a, miR133b, and miR206 [113]. When MPCs, i.e., mesenchymal progenitor cells isolated from traumatized muscle tissue of patients, were transfected with miRNA mimics encoding selected miRNAs and induced to undergo osteogenic differentiation, the most potent osteogenic inductors were identified for myomiRs—miR1 and miR206. Next, in vitro and in silico analyses allowed the identification of SOX9, which is involved, among others, in chondrocyte differentiation, as a candidate downstream target of miR1 and miR206 miRNAs in osteogenic differentiation. Investigation of the expression level of SOX9 in samples from patients developing HO showed that it is downregulated in samples obtained from patients developing patients, as compared to control [113]. Thus, SOX9 expression is modulated during the development and progression of HO.

It was documented that progenitors, residing in the interstitium of skeletal muscle and expressing the platelet-derived growth factor receptor α (PDGFRα+) marker, may participate in the formation of HO [190]. Recently, an in vitro study by Zhu et al. in which human PDGFRα+ muscle cells were induced to undergo osteogenic differentiation showed that miR19b-3p could be involved in this process, as its level increases as differentiation progresses. [191]. miR19b-3p acts by indirect induction of OCN, OPN/SPP1, and RUNX2 levels [191]. Furthermore, miR19b3p down-regulates phosphate and tension homolog deleted on chromosome ten (PTEN), which is involved in the regulation of bone formation by inhibiting the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, which was shown during osteogenic differentiation of stem cells [192, 193]. Transfection of PDGFRα+ muscle cells with miR19b3p mimic or miR19b3p inhibitor resulted in a decrease in PTEN mRNA when miR19b-3p was overexpressed and increased when miR19b3p was knocked down. Thus, miR19b 3p promotes PDGFRα+ muscle cell osteogenic differentiation of PDGFR+ muscle cells by inhibiting PTEN. Importantly, inhibition of miR19b3p inhibits osteogenesis of PDGFRα+ muscle cells, suggesting that miR9b3p could be a therapeutic target in future therapy against HO. Another molecule regulating the PTEN/PI3K/AKT signaling pathway during osteogenic differentiation of in vitro cultured human primary chondrocytes was miR181a/b-1. Zheng et al. showed that stable lentiviral overexpression of miR181a/b-1 in human chondrocytes enhanced osteogenesis in vitro [194]. Furthermore, PTEN expression was lower in human chondrocytes in which miR181a/b-1 was overexpressed, and consequently, PI3K/AKT signaling was increased [194]. These findings suggest that miR181a/b could also be a target in therapies for bone conditions such as fractures or HO. Another study by Qin et al. investigated miR-17-5p targeting the ankylosis protein homolog (ANKH), a gene associated with ankylosing spondylitis (AS). The miR-17-5p inhibitor reduces heterotopic bone formation in samples from the human hip joint capsule [195].

miRNA that targets ACVR1/ALK2

miR148 was described as a probable target for the development of therapeutic agents against FOP [196]. It directly targeted and downregulated ACVR1/ALK2 mRNA and protein in HeLa cells. Furthermore, in HeLa cells, miR148 downregulated the mRNA of the inhibitor of DNA binding -1, -2, and -3 (ID-1, -2, -3), suppressing the BMP signaling pathway [196]. Point mutations of the ACVR1/ALK2 gene and their constitutive activation of the BMP signaling pathway are observed in patients with FOP. Constitutively active ACVR1/ALK2 was also shown to cause endothelial–mesenchymal transition of endothelial cells, leading to FOP lesions. However, more studies are needed to examine the role of miR148 in FOP and other HO patients. Another miRNA targeting ACVR1/ALK2 mRNA is miR208a-3p [197]. However, its role in osteoblast differentiation was studied in the mouse hind limb unloading (HLU) model. The overexpression of miR-208a-3p was inhibited, while the silencing of miR-208a-3p with antagomiR-208a-3p promoted osteoblast differentiation [197].

lncRNA and heterotopic ossification

In vitro studies using human bone marrow mesenchymal stromal cells (hMSCs) showed that lncRNA could also be involved in osteogenesis and bone formation [198]. MSCs were also considered a source of cells responsible for HO formation [39]. The overexpression of lncRNA H19 promoted the osteogenic differentiation of hMSCs in vitro [198]. Furthermore, miR675 encoded by exon 1 of the lncRNA H19 also had an osteogenic effect in hMSC [198]. lncRNA H19 and miR675 downregulated TGFβ1 leading to inhibition of SMAD3 phosphorylation inhibition [198]. On the other hand, TGFβ1 was shown to inhibit osteogenic differentiation of hMSCs. Furthermore, lncRNA H19 and miR675 negatively regulated HDAC4/5, and thus increased the expression of osteoblast markers, such as RUNX2, expression [198]. Cells overexpressing lncRNA H19 transplanted subcutaneously more efficiently formed the bone. However, the role of lncRNA H19 or miR675 was not studied in HO patients or mice models of HO formation.

In vitro and in vivo studies using human adipose-derived stem cells (hASCs) showed that lncRNA MIAT (myocardial infarction-associated transcript), which plays an important role in signaling pathways such as Hippo, PI3K/AKT/c-MET, and WNT/β-catenin, is significantly downregulated in osteogenic differentiation of stem cells [199]. MIAT knockdown in hASC reverses the inhibition induced by tumor necrosis factor α (TNFα) of osteogenesis. However, its precise role and mechanism of action remained unknown [199]. Another factor that could be involved in HO formation is bromodomain-containing protein 4 (BRD4), that is, a regulator of gene expression involved in osteoclast differentiation. BRD4 belongs to the bromodomain and extraterminal (BET) protein family and participates in the organization of superenhancers and the regulation of oncogene expression. In the HO model, overexpression of BRD4 was associated with an increased level of mitotically associated lncRNA, i.e., Mancr [200], which role was first identified in invasive breast cancer [201]. The BRD4 acting through Mancr lncRNA increased the expression of RUNX2, OSX gene, and ALP encoding gene and, which, as a result, led to HO induction. In this study, a new BRD4-Mancr-RUNX2 pathway was identified, associated with HO signaling, was identified [200]. Nagasawa et al. hypothesized that Mancr activates RUNX2 expression by another ncRNA—miR-218 [202]. Furthermore, Liu et al. described the use of JQ1, a BRD4-specific antagonist that reduces HO formation [200]. Similarly, silencing Mancr inhibited osteogenesis [200]. Next, Hatzikotoulas et al. hypothesized that variation in the CAS20 locus that encodes lncRNA is responsible for susceptibility to HO in patients undergoing THA. Upon BMP2 causing osteogenic differentiation of MSCs, the expression of CASC20 was induced. CASC20 overexpression was related to RUNX2 and OSX activation and resulted in mineralized tissue formation [42]. The identified studies on ncRNA participating in the formation of HO are listed in Table 2.

Table 2 ncRNA participation in the formation of HO

Conclusions

Role of ncRNA in HO pathogenesis

The review sums up to date literature on the involvement of ncRNA in the processes that could underlay the HO (Fig. 3). The available data are limited and are based only on several research papers. In some of them, animal models of traumatic heterotopic ossification—Achilles tenotomy in mice [68, 200] or specific muscle/chondrocytes cell lines were used [191]. Other studies included profiling of ncRNA in humans, including post-traumatic HO [113, 173], patients after THA surgery [200] and NHO following CNS injury [189]. Of all published studies, the vast majority concerned only selected miRNA (miR-1, miR-19b-3p, miR-133a, miR-133b, miR-148a, miR-203, miR-206, miR 421, miR 433 and miR-630). Most of the ones that were previously identified by bioinformatic analysis and shown to target specific elements of the HO signaling network elements, e.g., ACVR1/ALK2 [196], BMP2 [180]. Other miRNA studies aimed at targets such as Slug, which is the regulator of endothelial–mesenchymal transition [177], OPN/SSP1 mRNA [189] or PTEN that controls osteogenic differentiation of muscle cells [90]. Only a few studies analyzed the role of lncRNA in the pathogenesis of HO (lncRNA MIAT, lncRNA Mancr, and lncRNA CASC20) [42, 199, 200]. Two studies proposed a different approach and described complex miRNA profiling and changes in trauma-induced HO [68, 113]. Importantly, it was shown which ncRNA could potentially serve as a biomarker of HO development and its severity. What is important, none of the specific ncRNA that was found to be significantly dysregulated in OPLL (miR-10a, miR-563, miR-199b, miR-182, miR-615, miR-132, lncR MALAT1, and lncR XIST) was investigated in other types of HO or its animal model [28].

Fig. 3
figure 3

Confirmed miRNA targets possibly involved in HO formation. miR-148a targets ACVR1 coding mRNA that results in a decrease in ACVR1 expression. ACVR1 is one of the BMPRs that acts as receptors for BMP. BMP binding to BMPRs results in activation of SMAD-dependent or SMAD-independent signaling pathways that leads to activation of transcription factors involved in osteogenesis, such as RUNX2 or DLX5. Other pathways potentially involved in HO formation are WNT, NF-κB or the HIF1 pathway. Activation of all these pathways leads to expression of transcription factors, such as RUNX2, DLX5, or OSX, which play a crucial role in bone formation, as well as BMPs. miR-203 is known to be a negative regulator of Runx2 translation, while miR-421 acts as a negative regulator of BMPs translation. The figure was created for this article; it is not based on any previously published image

Most studies evaluated miRNAs and their impact on bone morphogenesis signaling, including canonical BMP [180, 196] or WNT/β-catenin and MAPK pathways [68]. The expression of the osteogenesis master regulator RUNX2 [68, 113, 177, 191] or levels of well-known osteogenic biomarkers, i.e., OCN [68, 113, 177, 191], OPN/SSP1 [177, 189, 191], ALP [68, 113, 177, 191], and BSP [68] or BMP2 [180] were assessed to determine effect of the miRNAs investigated. Similarly, studies in HO identified lncRNA (MIAT, Mancr, and CASC20) that aim for OSX [42, 200] ALP [42, 199, 200], OCN [42, 199], RUNX2 [42, 199, 200] as their effectors. Until now, no ncRNA studies addressed recently described pathways involved in HO, such as HIF-1α, mTOR, or NF-κB [39].

In few identified studies, researchers successfully blocked osteogenesis or HO formation using miRNA mimics or shRNA (e.g., [177]). Other ncRNAs, i.e., siRNA against RUNX2, OSX, or SMAD, were shown to inhibit HO [203,204,205,206]. Thus, ncRNA and its intracellular regulations are a potential target for future HO therapy. Although, to date, none of the miRNA molecules are tested in human clinical trials to treat HO [207].

Future perspectives of ncRNA in HO

The roles of noncoding RNAs in the pathogenesis of HO remain largely unknown and undefined. Therefore, it is not clear would be the impact of such investigation on the progress in developing appropriate therapies. Each of the research works identifies different RNAs which could indicate that the exact regulatory role of ncRNA in HO remains unknown. Possible research directions for the future, providing such critical but still missing information, include the comparison of the expression profile in different types of HO and normal tissues rather than the comparison of binary interactions.

The therapeutic use of ncRNA in HO requires a design of the delivery method to target cells that ensures stability and safety. One of the challenges of ncRNA therapies is the method of delivery, which is handled by the use of viral (adenovirus, lentivirus) or non-viral vectors [208]. The delivery of viral vectors is efficient, but there is a risk of immunogenicity, toxicity, and carcinogenesis [208]. Therefore, constructs, such as locked nucleic acid [209], cholesterol-conjugated miRNA, lipid particles [210], or bacterially derived minicells, could be used [211]. Another issue is the stability of ncRNA, which could be improved by chemical modifications that prevent it from nuclease [208, 212]. To avoid the immune response, chemical modifications or small molecule inhibitors of ncRNA could be used [163]. To avoid off-target effects, ncRNA can be enriched with cell-specific ligands [208]. Compared to other drugs tested in FOP such as antibodies and small molecules, ncRNA has some potential therapeutic benefits as a therapeutic target. The half-life of ncRNA drugs may be very long, which means more patient-friendly doses [213]. In addition, unlike antibodies, the ncRNA can be transferred from cell to cell by the paracrine effect in exosomes, increasing the bioavailability of the drug in the affected tissue [214]. This review has presented that miRNA and lncRNA are intensively studied in the aspect of HO in both animal models and humans. However, given the complexity of this pathological process involving multiple pathways and possible effector cells in different HO types, the knowledge about the role of ncRNAs in the formation of HO is still very limited.

Availability of data and materials

Not applicable.

Abbreviations

ACVR1/ALK2:

Activin A receptor type 1/activin receptor-like kinase-2

AGO2:

Argonaute RISC catalytic component 2

AHO:

Albright osteodystrophy

AKT:

Protein kinase B

ALP :

Alkaline phosphatase

ANKH:

Ankylosis protein homolog

ASO:

Antisense oligonucleotides

AS:

Ankylosing spondylitis

BL:

Baseline

BMP:

Bone morphogenetic protein

BRD4:

Bromodomain-containing protein 4

BSP :

Bone sialoprotein

CNS:

Central nervous system

CD-RAP:

Cartilage-derived retinoic acid-sensitive protein

COX:

Cyclooxygenase

DAMP:

Damage-associated molecular pattern

DGCR8:

Di George syndrome critical-related gene 8

DLX5:

Distal-less homeobox 5

EMA:

European Medicines Agency

FOP:

Fibrodysplasia ossificans progressiva

FXR1:

Fragile X mental retardation-related protein 1

hASCs:

Human adipose-derived stem cells

HDAC4/5:

Histone deacetylase 4/5

HH:

Hedgehog

HIF-1α :

Hypoxia-inducible factor 1α

HLU:

Hindlimb unloading

HMGB1:

High-mobility group box 1

hMSCs:

Human bone marrow mesenchymal stromal cells

HO:

Heterotopic ossification

HOTL:

Heterotopic ossification of the tendon and ligament

HSP:

Heat shock protein

IL-1β:

Interleukin 1β

IL-6:

Interleukin 6

lncRNA:

Long noncoding RNA

LPS:

Lipopolysaccharide

Mancr:

Mitotically associated long noncoding RNA

MIAT :

Myocardial infarction-associated transcript

miRNA:

MicroRNA

MAPK:

Mitogen-activated protein kinase

MPCs :

Mesenchymal progenitor cells

MVECs:

Microvascular endothelial cells

mTOR:

Mammalian target of rapamycin

mTORC1:

Mammalian target of rapamycin protein complex-1

ncRNA:

Noncoding RNA

NF-κB:

Nuclear factor-κB

NHO:

Neurogenic heterotopic ossification

NSAIDs:

Nonsteroidal anti-inflammatory drugs

NT-3:

Neurotrophin-3

OCN:

Osteocalcin

OPLL:

Ossification of posterior longitudinal ligament of the spine

OPN/SSP1:

Osteopontin

OSM:

Oncostatin M

OSX:

Osterix

PAMP:

Pathogen-associated molecular pattern

PET:

Positron emission tomography

PI3K:

Phosphoinositide 3-kinase

POH:

Progressive osseous heteroplasia

POLII:

Polymerase II

PROMs:

Patient-reported outcome measures

pri-miRNA:

Primary miRNA transcript

PTEN:

Phosphate and tension homolog deleted on chromosome ten

RCT:

Randomized controlled trial

RISC:

RNA-induced silencing complex

ROM:

Range of motion

RT:

Radiotherapy

RUNX2:

Runt-related transcription factor 2

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

siRNA:

Small interfering RNA

snoRNA:

Small nucleolar RNA

snRNA:

Small nuclear RNA

SOX9:

SRY-box transcription factor 9

SP:

Substance P

TBI:

Traumatic brain injury

TGFβ:

Transforming growth factor β

THA:

Total hip arthroplasty

TLR:

Toll-like receptor

TNFα:

Tumor necrosis factor α

tRNAs:

Transfer RNA

WBCT:

Whole-body computed tomography

XPO5:

Exportin 5

References

  1. Meyers C, et al. Heterotopic ossification: a comprehensive review. JBMR Plus. 2019;3(4):e10172.

    Article  Google Scholar 

  2. Xu Y, et al. Heterotopic ossification: clinical features, basic researches, and mechanical stimulations. Front Cell Dev Biol. 2022. https://doi.org/10.3389/fcell.2022.770931.

    Article  Google Scholar 

  3. Nauth A, et al. Heterotopic ossification in orthopaedic trauma. J Orthop Trauma. 2012;26(12):684–8.

    Article  Google Scholar 

  4. Zhou L, et al. Heterotopic ossification after arthroscopic procedures: a scoping review of the literature. Orthop J Sports Med. 2022;10(1):23259671211060040.

    Article  Google Scholar 

  5. Rüdiger HA, et al. The impact of heterotopic ossification on self-reported outcomes after total hip arthroplasty using the direct anterior approach. J Bone Jt Surg Am. 2020;102(Suppl 2):91–8.

    Article  Google Scholar 

  6. Neal B, et al. Incidence of heterotopic bone formation after major hip surgery. ANZ J Surg. 2002;72(11):808–21.

    Article  Google Scholar 

  7. Pohl F, et al. The influence of heterotopic ossification on functional status of hip joint following total hip arthroplasty. Strahlenther Onkol. 2005;181(8):529–33.

    Article  Google Scholar 

  8. Gkiatas I, et al. Heterotopic ossification negatively influences range of motion after revision total knee arthroplasty. J Arthroplast. 2021;36(8):2907–12.

    Article  Google Scholar 

  9. Skorochod R, Nesher G, Gronovich Y. Heterotopic ossification in patients with burns: a systematic literature review. Curr Phys Med Rehabil Rep. 2022. https://doi.org/10.1007/s40141-022-00356-5.

    Article  Google Scholar 

  10. Hong CC, et al. Clinically relevant heterotopic ossification after elbow fracture surgery: a risk factors study. Orthop Traumatol Surg Res. 2015;101(2):209–13.

    Article  CAS  Google Scholar 

  11. Ahrengart L, Lindgren U. Functional significance of heterotopic bone formation after total hip arthroplasty. J Arthroplasty. 1989;4(2):125–31.

    Article  CAS  Google Scholar 

  12. Wong KR, et al. Neurological heterotopic ossification: novel mechanisms, prognostic biomarkers and prophylactic therapies. Bone Res. 2020;8(1):42.

    Article  CAS  Google Scholar 

  13. Johns JS, et al. Impact of clinically significant heterotopic ossification on functional outcome after traumatic brain injury. J Head Trauma Rehabil. 1999;14(3):269–76.

    Article  CAS  Google Scholar 

  14. Taly AB, et al. Neurogenic heterotopic ossification: a diagnostic and therapeutic challenge in neurorehabilitation. Neurol India. 2001;49(1):37–40.

    CAS  Google Scholar 

  15. Meyer C, et al. Heterotopic ossification in COVID-19: a series of 4 cases. Ann Phys Rehabil Med. 2020;63(6):565–7.

    Article  CAS  Google Scholar 

  16. Mezghani S, et al. Heterotopic ossification and COVID 19: imaging analysis of ten consecutive cases. Eur J Radiol. 2022;152:110336.

    Article  Google Scholar 

  17. Zhang Q, et al. Heterotopic ossification of tendon and ligament. J Cell Mol Med. 2020;24(10):5428–37.

    Article  Google Scholar 

  18. McCarthy EF, Sundaram M. Heterotopic ossification: a review. Skeletal Radiol. 2005;34(10):609–19.

    Article  CAS  Google Scholar 

  19. Vanden Bossche L, Vanderstraeten G. Heterotopic ossification: a review. J Rehabil Med. 2005;37(3):129–36.

    Article  Google Scholar 

  20. Abiola R, Rubery P, Mesfin A. Ossification of the posterior longitudinal ligament: etiology, diagnosis, and outcomes of nonoperative and operative management. Glob Spine J. 2016;6(2):195–204.

    Article  Google Scholar 

  21. Oliva F, Via AG, Maffulli N. Calcific tendinopathy of the rotator cuff tendons. Sports Med Arthrosc Rev. 2011;19(3):237–43.

    Article  Google Scholar 

  22. Le HV, et al. Ossification of the posterior longitudinal ligament: pathophysiology, diagnosis, and management. J Am Acad Orthop Surg. 2022. https://doi.org/10.5435/JAAOS-D-22-00049.

    Article  Google Scholar 

  23. Maeda S, et al. Functional impact of human collagen α2 (XI) gene polymorphism in pathogenesis of ossification of the posterior longitudinal ligament of the spine. J Bone Miner Res. 2001;16(5):948–57.

    Article  CAS  Google Scholar 

  24. Kong Q, et al. COL6A1 polymorphisms associated with ossification of the ligamentum flavum and ossification of the posterior longitudinal ligament. Spine. 2007;32(25):2834–8.

    Article  Google Scholar 

  25. Okamoto K, et al. Dietary habits and risk of ossification of the posterior longitudinal ligaments of the spine (OPLL); findings from a case-control study in Japan*. J Bone Miner Metab. 2004;22(6):612–7.

    Article  Google Scholar 

  26. Hirai T, et al. Prevalence and distribution of ossified lesions in the whole spine of patients with cervical ossification of the posterior longitudinal ligament a multicenter study (JOSL CT study). PLoS ONE. 2016;11(8):e0160117.

    Article  Google Scholar 

  27. Endo T, et al. Close association between non-alcoholic fatty liver disease and ossification of the posterior longitudinal ligament of the spine. Sci Rep. 2021;11(1):17412.

    Article  CAS  Google Scholar 

  28. Yuan X, Shi L, Chen Y. Non-coding RNAs in ossification of spinal ligament. Eur Spine J. 2021;30(4):801–8.

    Article  Google Scholar 

  29. Darrieutort-Laffite C, Blanchard F, Le Goff B. Calcific tendonitis of the rotator cuff: From formation to resorption. Jt Bone Spine. 2018;85(6):687–92.

    Article  Google Scholar 

  30. Uhthoff HK. Calcifying tendinitis, an active cell-mediated calcification. Virchows Arch A Pathol Anat Histol. 1975;366(1):51–8.

    Article  CAS  Google Scholar 

  31. Cappato S, et al. Genetic and acquired heterotopic ossification: a translational tale of mice and men. Biomedicines. 2020;8(12):611.

    Article  CAS  Google Scholar 

  32. De Brasi D, et al. Fibrodysplasia ossificans progressiva: a challenging diagnosis. Genes (Basel). 2021;12(8):1187.

    Article  Google Scholar 

  33. Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med. 2013;19(2):179–92.

    Article  CAS  Google Scholar 

  34. Liljesthröm M, Pignolo RJ, Kaplan FS. Epidemiology of the global fibrodysplasia ossificans progressiva (FOP) community. J Rare Dis Res Treat. 2020;5(2):31–6.

    Article  Google Scholar 

  35. Kapoor S, et al. Albright’s hereditary osteodystrophy. Indian J Pediatr. 2006;73:153–6.

    Article  Google Scholar 

  36. Mantovani G, et al. Diagnosis and management of pseudohypoparathyroidism and related disorders: first international consensus statement. Nat Rev Endocrinol. 2018;14(8):476–500.

    Article  CAS  Google Scholar 

  37. Rocke DM, et al. Age- and joint-specific risk of initial heterotopic ossification in patients who have fibrodysplasia ossificans progressiva. Clin Orthop Relat Res. 1994;301:243–8.

    Article  Google Scholar 

  38. Peng K, et al. Longitudinal evaluation of pain, flare-up, and emotional health in fibrodysplasia ossificans progressiva: analyses of the international FOP registry. JBMR Plus. 2019;3(8):e10181.

    Article  Google Scholar 

  39. Pulik Ł, et al. The survey of cells responsible for heterotopic ossification development in skeletal muscles-human and mouse models. Cells. 2020;9(6):1324.

    Article  CAS  Google Scholar 

  40. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.

    Article  Google Scholar 

  41. Mitchell EJ, et al. The genetics of heterotopic ossification: insight into the bone remodeling pathway. J Orthop Trauma. 2010;24(9):530–3.

    Article  Google Scholar 

  42. Hatzikotoulas K et al. Genome-wide association and functional analyses identify CASC20 and KIF26B as target loci in heterotopic ossification. bioRxiv, 2019; 845958.

  43. Yang Z, et al. Potential genes and pathways associated with heterotopic ossification derived from analyses of gene expression profiles. J Orthop Surg Res. 2021;16(1):499.

    Article  Google Scholar 

  44. Logan NJ, et al. Demethylation of ITGAV accelerates osteogenic differentiation in a blast-induced heterotopic ossification in vitro cell culture model. Bone. 2018;117:149–60.

    Article  CAS  Google Scholar 

  45. Edsberg LE, et al. A survey of proteomic biomarkers for heterotopic ossification in blood serum. J Orthop Surg Res. 2017;12(1):69–69.

    Article  Google Scholar 

  46. Wei Z, et al. Comparative proteomic analysis identifies differentially expressed proteins and reveals potential mechanisms of traumatic heterotopic ossification progression. J Orthop Transl. 2022;34:42–59.

    Google Scholar 

  47. Wang L, et al. Could heterotopic ossification be prevented by varying dietary n-3/n-6 polyunsaturated fatty acid ratio: a novel perspective to its treatment? Med Hypotheses. 2013;80(1):57–60.

    Article  CAS  Google Scholar 

  48. Lees-Shepard JB, Goldhamer DJ. Stem cells and heterotopic ossification: lessons from animal models. Bone. 2018;109:178–86.

    Article  Google Scholar 

  49. Lounev VY, et al. Identification of progenitor cells that contribute to heterotopic skeletogenesis. J Bone Jt Surg Am Vol. 2009;91(3):652–63.

    Article  Google Scholar 

  50. Huang Y, et al. Macrophages in heterotopic ossification: from mechanisms to therapy. npj Regen Med. 2021;6(1):70.

    Article  Google Scholar 

  51. Convente MR, et al. The immunological contribution to heterotopic ossification disorders. Curr Osteoporos Rep. 2015;13(2):116–24.

    Article  Google Scholar 

  52. Tseng HW, et al. Interleukin-1 is overexpressed in injured muscles following spinal cord injury and promotes neurogenic heterotopic ossification. J Bone Miner Res. 2022;37(3):531–46.

    Article  CAS  Google Scholar 

  53. Marsell R, Einhorn TA. The biology of fracture healing. Injury. 2011;42(6):551–5.

    Article  Google Scholar 

  54. Mackie EJ, et al. Endochondral ossification: how cartilage is converted into bone in the developing skeleton. Int J Biochem Cell Biol. 2008;40(1):46–62.

    Article  CAS  Google Scholar 

  55. Foley KL, et al. Histopathology of periarticular non-hereditary heterotopic ossification. Bone. 2018;109:65–70.

    Article  CAS  Google Scholar 

  56. Culbert AL, et al. Alk2 regulates early chondrogenic fate in fibrodysplasia ossificans progressiva heterotopic endochondral ossification. Stem Cells. 2014;32(5):1289–300.

    Article  CAS  Google Scholar 

  57. Lindborg CM, et al. Cartilage-derived retinoic acid-sensitive protein (CD-RAP): a stage-specific biomarker of heterotopic endochondral ossification (HEO) in fibrodysplasia ossificans progressiva (FOP). Bone. 2018;109:153–7.

    Article  CAS  Google Scholar 

  58. Kaplan FS, et al. The histopathology of fibrodysplasia ossificans progressive. An endochondral process. J Bone Jt Surg Am. 1993;75(2):220–30.

    Article  CAS  Google Scholar 

  59. Kaplan FS, Shore EM. Progressive osseous heteroplasia. J Bone Miner Res. 2000;15(11):2084–94.

    Article  CAS  Google Scholar 

  60. DeSesso JM, Scialli AR. Bone development in laboratory mammals used in developmental toxicity studies. Birth Defects Res. 2018;110(15):1157–87.

    Article  CAS  Google Scholar 

  61. Xian CJ, et al. Intramembranous ossification mechanism for bone bridge formation at the growth plate cartilage injury site. J Orthop Res. 2004;22(2):417–26.

    Article  Google Scholar 

  62. Barruet E, et al. NF-κB/MAPK activation underlies ACVR1-mediated inflammation in human heterotopic ossification. JCI Insight. 2018;3(22):e122958.

    Article  Google Scholar 

  63. Huang Y, Wang X, Lin H. The hypoxic microenvironment: a driving force for heterotopic ossification progression. Cell Commun Signal. 2020;18(1):20.

    Article  Google Scholar 

  64. Wu J, et al. BMP and mTOR signaling in heterotopic ossification: does their crosstalk provide therapeutic opportunities? J Cell Biochem. 2019;120(8):12108–22.

    Article  CAS  Google Scholar 

  65. Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci. 2009;122(20):3589–94.

    Article  CAS  Google Scholar 

  66. Kan C, et al. Conserved signaling pathways underlying heterotopic ossification. Bone. 2018;109:43–8.

    Article  CAS  Google Scholar 

  67. Wang S, et al. Identification of the biomarkers and pathological process of heterotopic ossification: weighted gene co-expression network analysis. Front Endocrinol. 2020;11:581768–581768.

    Article  Google Scholar 

  68. Tu B, et al. miR-203 inhibits the traumatic heterotopic ossification by targeting Runx2. Cell Death Dis. 2016;7(10):e2436–e2436.

    Article  CAS  Google Scholar 

  69. Migliorini F, et al. NSAIDs for prophylaxis for heterotopic ossification after total hip arthroplasty: a Bayesian network meta-analysis. Calcif Tissue Int. 2021;108(2):196–206.

    Article  CAS  Google Scholar 

  70. Łęgosz P, et al. Challenges of heterotopic ossification-molecular background and current treatment strategies. Clin Exp Pharmacol Physiol. 2018;45(12):1229–35.

    Article  Google Scholar 

  71. Takada Y, et al. Nonsteroidal anti-inflammatory agents differ in their ability to suppress NF-kappaB activation, inhibition of expression of cyclooxygenase-2 and cyclin D1, and abrogation of tumor cell proliferation. Oncogene. 2004;23(57):9247–58.

    Article  CAS  Google Scholar 

  72. Pakos EE, Ioannidis JP. Radiotherapy versus nonsteroidal anti-inflammatory drugs for the prevention of heterotopic ossification after major hip procedures: a meta-analysis of randomized trials. Int J Radiat Oncol Biol Phys. 2004;60(3):888–95.

    Article  CAS  Google Scholar 

  73. Patel AB, et al. Radiation therapy prophylaxis for heterotopic ossification in non-hip sites. J Cancer Ther. 2018;9(1):1–8.

    Article  Google Scholar 

  74. Sautter-Bihl ML, Liebermeister E, Nanassy A. Radiotherapy as a local treatment option for heterotopic ossifications in patients with spinal cord injury. Spinal Cord. 2000;38(1):33–6.

    Article  CAS  Google Scholar 

  75. Łęgosz P, et al. Heterotopic ossification: a challenging complication of total hip arthroplasty: risk factors, diagnosis, prophylaxis, and treatment. Biomed Res Int. 2019;2019:3860142.

    Article  Google Scholar 

  76. Milakovic M, et al. Radiotherapy for the prophylaxis of heterotopic ossification: a systematic review and meta-analysis of randomized controlled trials. Radiother Oncol. 2015;116(1):4–9.

    Article  Google Scholar 

  77. Hu Z-H, et al. Radiotherapy for the prophylaxis of heterotopic ossification after total hip arthroplasty: a systematic review and meta-analysis of randomized controlled trails. Med Dosim. 2021;46(1):65–73.

    Article  Google Scholar 

  78. Galietta E, et al. Prophylactic radiotherapy of hip heterotopic ossification: a narrative mini review. In Vivo. 2022;36(2):533–42.

    Article  Google Scholar 

  79. Rosenberg DM, et al. Radiation-induced sarcoma after heterotopic ossification prophylaxis: a case report. JBJS Case Connect. 2019;9(4):e0146.

    Article  Google Scholar 

  80. Farris MK, et al. Osteosarcoma following single fraction radiation prophylaxis for heterotopic ossification. Radiat Oncol. 2012;7(1):1–6.

    Article  Google Scholar 

  81. Mourad WF, et al. Radiation-induced sarcoma following radiation prophylaxis of heterotopic ossification. Pract Radiat Oncol. 2012;2(2):151–4.

    Article  Google Scholar 

  82. Hanna M, Farid YR, Finn HA. Low-dose preoperative unshielded radiation is effective in heterotopic ossification prophylaxis and does not affect porous fixation in total hip arthroplasty at 2 years minimum follow-up: a radiographic study. JAAOS: J Am Acad Orthop Surg. 2022;30(5):223–8.

    Article  Google Scholar 

  83. Shapira J, et al. Efficacy of NSAIDs versus radiotherapy for heterotopic ossification prophylaxis following total hip arthroplasty in high-risk patients: a systematic review and meta-analysis. HIP Int. 2021;32(5):576–90.

    Article  Google Scholar 

  84. Vavken P, Dorotka R. Economic evaluation of NSAID and radiation to prevent heterotopic ossification after hip surgery. Arch Orthop Trauma Surg. 2011;131(9):1309–15.

    Article  Google Scholar 

  85. Wang Y, et al. Irradiation alters the differentiation potential of bone marrow mesenchymal stem cells. Mol Med Rep. 2016;13(1):213–23.

    Article  CAS  Google Scholar 

  86. Felix-Ilemhenbhio F, et al. Pathophysiology and emerging molecular therapeutic targets in heterotopic ossification. Int J Mol Sci. 2022;23(13):6983.

    Article  CAS  Google Scholar 

  87. Vanhoutte F, et al. Pharmacokinetics and pharmacodynamics of garetosmab (Anti-Activin A): results from a first-in-human phase 1 study. J Clin Pharmacol. 2020;60(11):1424–31.

    Article  CAS  Google Scholar 

  88. Dahir KM, et al. Garetosmab Reduces flare-ups in patients with fibrodysplasia ossificans progressiva. J Endocr Soc. 2021;5(Supplement_1):A251–2.

    Article  Google Scholar 

  89. Ventura F, et al. Challenges and opportunities for drug repositioning in fibrodysplasia ossificans progressiva. Biomedicines. 2021;9(2):213.

    Article  CAS  Google Scholar 

  90. The Examination of Safety and Efficacy of Garetosmab Versus Placebo Administered Intravenously (IV) in Adult Participants With Fibrodysplasia Ossificans Progressiva (OPTIMA). ClinicalTrials.gov identifier: NCT05394116. Updated May 27, 2022. https://ClinicalTrials.gov/show/NCT05394116. Accessed 23 July 2022.

  91. Study of Single-Ascending Doses of DS-6016a in Healthy Japanese. ClinicalTrials.gov identifier: NCT04818398. Updated January 10, 2022. https://ClinicalTrials.gov/show/NCT04818398. Accessed 23 July 2022.

  92. Rooney L, Jones C. Recent advances in ALK2 inhibitors. ACS Omega. 2021;6(32):20729–34.

    Article  CAS  Google Scholar 

  93. Ordonez C, et al. Administration of KER-047, a novel ALK2 Inhibitor, elicited robust and sustained increases in serum iron in healthy participants. Blood. 2020;136(Supplement 1):13–13.

    Article  Google Scholar 

  94. Sabir AH, Cole T. The evolving therapeutic landscape of genetic skeletal disorders. Orphanet J Rare Dis. 2019;14(1):300.

    Article  Google Scholar 

  95. Study to Assess the Efficacy and Safety of 2 Dosage Regimens of Oral IPN60130 for the Treatment of Fibrodysplasia Ossificans Progressiva (FOP) (FALKON). ClinicalTrials.gov identifier: NCT05039515. Updated July 6, 2022. https://ClinicalTrials.gov/show/NCT05039515. Accessed 23 July 2022.

  96. To Assess the Efficacy, Safety, and Tolerability of INCB000928 in Participants With Fibrodysplasia Ossificans Progressiva (PROGRESS). ClinicalTrials.gov identifier: NCT05090891. Updated June 13, 2022. Available from: https://ClinicalTrials.gov/show/NCT05090891. Accessed 23 July 2022.

  97. Calpe S, et al. Comparison of newly developed anti-bone morphogenetic protein 4 llama-derived antibodies with commercially available BMP4 inhibitors. MAbs. 2016;8(4):678–88.

    Article  CAS  Google Scholar 

  98. Meng X, Wang H, Hao J, Recent progress in drug development for fibrodysplasia ossificans progressiva. Mol Cell Biochem 2022.

  99. Mundy C, et al. Activin A promotes the development of acquired heterotopic ossification and is an effective target for disease attenuation in mice. Sci Signal. 2021;14(669):536.

    Article  Google Scholar 

  100. Saracatinib Trial TO Prevent FOP (STOPFOP). ClinicalTrials.gov identifier: NCT04307953. Updated July 22, 2022. https://ClinicalTrials.gov/show/NCT04307953. Accessed 23 July 2022.

  101. Kitoh H. Clinical aspects and current therapeutic approaches for FOP. Biomedicines. 2020;8(9):325.

    Article  CAS  Google Scholar 

  102. Williams E, et al. Saracatinib is an efficacious clinical candidate for fibrodysplasia ossificans progressiva. JCI Insight. 2021;6(8):e95042.

    Article  Google Scholar 

  103. Hino K, et al. Activin-A enhances mTOR signaling to promote aberrant chondrogenesis in fibrodysplasia ossificans progressiva. J Clin Investig. 2017;127:3339–52.

    Article  Google Scholar 

  104. Maekawa H, et al. Prophylactic treatment of rapamycin ameliorates naturally developing and episode -induced heterotopic ossification in mice expressing human mutant ACVR1. Orphanet J Rare Dis. 2020;15(1):122.

    Article  Google Scholar 

  105. Multicenter randomized double-blind comparison test followed by open-label continuous administration test of NPC-12T for Fibrodysplasia Ossificans Progressiva. UMIN-CTR identifier: UMIN000028429. Updated February 2, 2022. https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr_view.cgi?recptno=R000032495. Accessed 23 July 2022.

  106. Hind M, Stinchcombe S. Palovarotene, a novel retinoic acid receptor gamma agonist for the treatment of emphysema. Curr Opin Investig Drugs. 2009;10(11):1243–50.

    CAS  Google Scholar 

  107. Shimono K, et al. Potent inhibition of heterotopic ossification by nuclear retinoic acid receptor-γ agonists. Nat Med. 2011;17(4):454–60.

    Article  CAS  Google Scholar 

  108. Huang J, et al. Palovarotene inhibits the NF-κB signalling pathway to prevent heterotopic ossification. Clin Exp Pharmacol Physiol. 2022;49(8):881–92.

    Article  CAS  Google Scholar 

  109. Huang J, et al. Palovarotene can attenuate heterotopic ossification induced by tendon stem cells by downregulating the synergistic effects of smad and NF-<i>κ</i>B signaling pathway following stimulation of the inflammatory microenvironment. Stem Cells Int. 2022;2022:1560943.

    Article  Google Scholar 

  110. An Efficacy and Safety Study of Palovarotene for the Treatment of Fibrodysplasia Ossificans Progressiva (MOVE). ClinicalTrials.gov identifier: NCT03312634. Updated March 15, 2022. https://ClinicalTrials.gov/show/NCT03312634. Accessed 23 July 2022.

  111. Jin W, et al. Engineered osteoclasts as living treatment materials for heterotopic ossification therapy. Nat Commun. 2021;12(1):6327.

    Article  CAS  Google Scholar 

  112. Hoy SM. Palovarotene: first approval. Drugs. 2022;82(6):711–6.

    Article  CAS  Google Scholar 

  113. de Vasconcellos JF, et al. A microRNA signature for impaired wound-healing and ectopic bone formation in humans. J Bone Jt Surg Am. 2020;102(21):1891–9.

    Article  Google Scholar 

  114. An Open-Label Extension Study of Palovarotene Treatment in Fibrodysplasia Ossificans Progressiva (FOP). ClinicalTrials.gov identifier: NCT02279095. Updated February 17, 2022. https://ClinicalTrials.gov/show/NCT02279095. Accessed 23 July 2022/

  115. An Open-Label Extension Study of Palovarotene to Prevent Heterotopic Ossification in People With Fibrodysplasia Ossificans Progressiva (FOP) in France. ClinicalTrials.gov identifier: NCT02979769. Updated February 17, 2022. https://ClinicalTrials.gov/show/NCT02979769. Accessed 23 July 2022.

  116. A Rollover Study to Further Evaluate the Safety and Efficacy of Palovarotene Capsules in Male and Female Participants Aged ≥14 Years With Fibrodysplasia Ossificans Progressiva (FOP) Who Have Completed the Relevant Parent Studies (PIVOINE). ClinicalTrials.gov identifier: NCT05027802. Updated July 1, 2022. https://ClinicalTrials.gov/show/NCT05027802. Accessed 23 July 2022.

  117. Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell. 2014;157(1):77–94.

    Article  CAS  Google Scholar 

  118. Hombach S, Kretz M. Non-coding RNAs: classification, biology and functioning. Adv Exp Med Biol. 2016;937:3–17.

    Article  CAS  Google Scholar 

  119. Zhang S, et al. The risks of miRNA therapeutics: In a drug target perspective. Drug Des Dev Ther. 2021;15:721–33.

    Article  Google Scholar 

  120. You C, et al. ODNA: a manually curated database of noncoding RNAs associated with orthopedics. Database (Oxford). 2019. https://doi.org/10.1093/database/baz126.

    Article  Google Scholar 

  121. Bartel DP. Metazoan MicroRNAs. Cell. 2018;173(1):20–51.

    Article  CAS  Google Scholar 

  122. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.

    Article  CAS  Google Scholar 

  123. O’Brien J, et al. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol. 2018;9:402.

    Article  CAS  Google Scholar 

  124. Dexheimer PJ, Cochella L. MicroRNAs: from mechanism to organism. Front Cell Dev Biol. 2020;8:409.

    Article  Google Scholar 

  125. Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. 2019;20(1):21–37.

    Article  CAS  Google Scholar 

  126. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15(8):509–24.

    Article  CAS  Google Scholar 

  127. Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16(7):421–33.

    Article  CAS  Google Scholar 

  128. Bukhari SIA, et al. A specialized mechanism of translation mediated by FXR1a-associated microRNP in cellular quiescence. Mol Cell. 2016;61(5):760–73.

    Article  CAS  Google Scholar 

  129. Truesdell SS, et al. MicroRNA-mediated mRNA translation activation in quiescent cells and oocytes involves recruitment of a nuclear microRNP. Sci Rep. 2012;2(1):842.

    Article  CAS  Google Scholar 

  130. Ørom UA, Nielsen FC, Lund AH. MicroRNA-10a binds the 5′UTR of ribosomal protein mRNAs and enhances their translation. Mol Cell. 2008;30(4):460–71.

    Article  Google Scholar 

  131. Gregory RI, et al. The microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432(7014):235–40.

    Article  CAS  Google Scholar 

  132. Denli AM, et al. Processing of primary microRNAs by the Microprocessor complex. Nature. 2004;432(7014):231–5.

    Article  CAS  Google Scholar 

  133. Lee Y, et al. The nuclear RNase III drosha initiates microRNA processing. Nature. 2003;425(6956):415–9.

    Article  CAS  Google Scholar 

  134. Han J, et al. The drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004;18(24):3016–27.

    Article  CAS  Google Scholar 

  135. Stavast CJ, Erkeland SJ. The non-canonical aspects of microRNAs: many roads to gene regulation. Cells. 2019;8(11):1465.

    Article  CAS  Google Scholar 

  136. Ruby JG, Jan CH, Bartel DP. Intronic microRNA precursors that bypass Drosha processing. Nature. 2007;448(7149):83–6.

    Article  CAS  Google Scholar 

  137. Okamura K, et al. The mirtron pathway generates microRNA-class regulatory RNAs in drosophila. Cell. 2007;130(1):89–100.

    Article  CAS  Google Scholar 

  138. Berezikov E, et al. Mammalian mirtron genes. Mol Cell. 2007;28(2):328–36.

    Article  CAS  Google Scholar 

  139. Okada C, et al. A high-resolution structure of the pre-microRNA nuclear export machinery. Science. 2009;326(5957):1275–9.

    Article  CAS  Google Scholar 

  140. Lund E, et al. Nuclear export of microRNA precursors. Science. 2004;303(5654):95–8.

    Article  CAS  Google Scholar 

  141. Bohnsack MT, Czaplinski K, Gorlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA (New York, NY). 2004;10(2):185–91.

    Article  CAS  Google Scholar 

  142. Guo L, Lu Z. The fate of miRNA* strand through evolutionary analysis: implication for degradation as merely carrier strand or potential regulatory molecule? PLoS ONE. 2010;5(6):e11387.

    Article  Google Scholar 

  143. Yang J-S, et al. Widespread regulatory activity of vertebrate microRNA* species. RNA (New York, NY). 2011;17(2):312–26.

    Article  CAS  Google Scholar 

  144. Yang X, et al. Both mature miR-17-5p and passenger strand miR-17-3p target TIMP3 and induce prostate tumor growth and invasion. Nucl Acids Res. 2013;41(21):9688–704.

    Article  CAS  Google Scholar 

  145. Pratt AJ, MacRae IJ. The RNA-induced silencing complex: a versatile gene-silencing machine. J Biol Chem. 2009;284(27):17897–901.

    Article  CAS  Google Scholar 

  146. Hutvágner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002;297(5589):2056–60.

    Article  Google Scholar 

  147. Hammond SM, et al. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature. 2000;404(6775):293–6.

    Article  CAS  Google Scholar 

  148. Wang Y, et al. Structure of an argonaute silencing complex with a seed-containing guide DNA and target RNA duplex. Nature. 2008;456(7224):921–6.

    Article  CAS  Google Scholar 

  149. Huntzinger E, Izaurralde E. Gene silencing by microRNAs: contributions of translational repression and mRNA decay. Nat Rev Genet. 2011;12(2):99–110.

    Article  CAS  Google Scholar 

  150. Wee LM, et al. Argonaute divides Its RNA guide into domains with distinct functions and RNA-binding properties. Cell. 2012;151(5):1055–67.

    Article  CAS  Google Scholar 

  151. Israeli D, et al. Circulating miRNAs are generic and versatile therapeutic monitoring biomarkers in muscular dystrophies. Sci Rep. 2016;6(1):28097.

    Article  CAS  Google Scholar 

  152. Cacchiarelli D, et al. miRNAs as serum biomarkers for duchenne muscular dystrophy. EMBO Mol Med. 2011;3(5):258–65.

    Article  CAS  Google Scholar 

  153. Vignier N, et al. Distinctive serum miRNA profile in mouse models of striated muscular pathologies. PLoS ONE. 2013;8(2):e55281.

    Article  CAS  Google Scholar 

  154. McHugh CA, et al. The xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature. 2015;521(7551):232–6.

    Article  CAS  Google Scholar 

  155. Wang X, et al. Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature. 2008;454(7200):126–30.

    Article  CAS  Google Scholar 

  156. Wang KC, et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 2011;472(7341):120–4.

    Article  CAS  Google Scholar 

  157. Clemson CM, et al. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol Cell. 2009;33(6):717–26.

    Article  CAS  Google Scholar 

  158. Atianand MK, et al. A long noncoding RNA lincRNA-EPS Acts as a transcriptional brake to restrain inflammation. Cell. 2016;165(7):1672–85.

    Article  CAS  Google Scholar 

  159. Martianov I, et al. Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript. Nature. 2007;445(7128):666–70.

    Article  CAS  Google Scholar 

  160. Feng J, et al. The Evf-2 noncoding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as a Dlx-2 transcriptional coactivator. Genes Dev. 2006;20(11):1470–84.

    Article  CAS  Google Scholar 

  161. Wang G-Q, et al. Sirt1 AS lncRNA interacts with its mRNA to inhibit muscle formation by attenuating function of miR-34a. Sci Rep. 2016;6(1):21865.

    Article  CAS  Google Scholar 

  162. Cesana M, et al. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 2011;147(2):358–69.

    Article  CAS  Google Scholar 

  163. Winkle M, et al. Noncoding RNA therapeutics: challenges and potential solutions. Nat Rev Drug Discov. 2021;20(8):629–51.

    Article  CAS  Google Scholar 

  164. Chen B, et al. Targeting non-coding RNAs to overcome cancer therapy resistance. Signal Transduct Target Ther. 2022;7(1):121.

    Article  CAS  Google Scholar 

  165. Mercer TR, Munro T, Mattick JS. The potential of long noncoding RNA therapies. Trends Pharmacol Sci. 2022;43(4):269–80.

    Article  CAS  Google Scholar 

  166. Kazezian Z, Bull AMJ. A review of the biomarkers and in vivo models for the diagnosis and treatment of heterotopic ossification following blast and trauma-induced injuries. Bone. 2021;143:115765.

    Article  Google Scholar 

  167. Łęgosz P, et al. The use of type I collagen cross-linked C-telopeptide (CTX-1) as a biomarker associated with the formation of periprosthetic ossifications following total hip joint arthroplasty. Ann Clin Lab Sci. 2018;48(2):183–90.

    Google Scholar 

  168. Sharma GG, et al. Non-coding RNA biomarkers in pancreatic ductal adenocarcinoma. Semin Cancer Biol. 2021;75:153–68.

    Article  CAS  Google Scholar 

  169. Gong K, et al. Comprehensive analysis of lncRNA biomarkers in kidney renal clear cell carcinoma by lncRNA-mediated ceRNA network. PLoS ONE. 2021;16(6):e0252452.

    Article  CAS  Google Scholar 

  170. Hu Y, et al. Comprehensive analysis of lncRNA-mRNAs co-expression network identifies potential lncRNA biomarkers in cutaneous squamous cell carcinoma. BMC Genom. 2022;23(1):274.

    Article  CAS  Google Scholar 

  171. Joilin G, et al. An overview of MicroRNAs as biomarkers of ALS. Front Neurol. 2019;10:186.

    Article  Google Scholar 

  172. Borga C, Meeran SM, Fassan M. Non-coding RNAs, a real next-gen class of biomarkers? Noncoding RNA Res. 2019;4(3):80–1.

    Article  Google Scholar 

  173. Ji Y et al. Finding Molecular Targets of Heterotopic Ossification Following War Trauma using microRNA. in ORS Annual Meeting. 2012.

  174. Chauhan N, et al. miR-205: a potential biomedicine for cancer therapy. Cells. 2020;9(9):1957.

    Article  CAS  Google Scholar 

  175. Vychytilova-Faltejskova P, Slaby O. MicroRNA-215: from biology to theranostic applications. Mol Aspects Med. 2019;70:72–89.

    Article  CAS  Google Scholar 

  176. McCarthy JJ. The MyomiR network in skeletal muscle plasticity. Exerc Sport Sci Rev. 2011;39(3):150–4.

    Article  Google Scholar 

  177. Sun Y, et al. MiR-630 inhibits endothelial-mesenchymal transition by targeting slug in traumatic heterotopic ossification. Sci Rep. 2016;6(1):22729.

    Article  CAS  Google Scholar 

  178. Jiang Z, et al. Increased expression of miR-421 in human gastric carcinoma and its clinical association. J Gastroenterol. 2010;45(1):17–23.

    Article  CAS  Google Scholar 

  179. Zhou H, et al. MiR-421 is a functional marker of circulating tumor cells in gastric cancer patients. Biomarkers. 2012;17(2):104–10.

    Article  CAS  Google Scholar 

  180. Ju C, et al. Regulatory effect of miR-421 on humeral fracture and heterotopic ossification in elderly patients. Exp Ther Med. 2019;17(3):1903–11.

    CAS  Google Scholar 

  181. Tao L, et al. Crucial role of miR-433 in regulating cardiac fibrosis. Theranostics. 2016;6(12):2068–83.

    Article  CAS  Google Scholar 

  182. Zhang T, et al. miR-433 inhibits breast cancer cell growth via the MAPK signaling pathway by targeting Rap1a. Int J Biol Sci. 2018;14(6):622–32.

    Article  CAS  Google Scholar 

  183. Shi Q, et al. MiR-433-3p inhibits proliferation and invasion of esophageal squamous cell carcinoma by targeting GRB2. Cell Physiol Biochem. 2018;46(5):2187–96.

    Article  CAS  Google Scholar 

  184. Sun S, et al. MiR-433-3p suppresses cell growth and enhances chemosensitivity by targeting CREB in human glioma. Oncotarget. 2017;8(3):5057–68.

    Article  Google Scholar 

  185. Jiang K, Teng GD, Chen YQ. 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(10):e3216.

    Article  CAS  Google Scholar 

  186. Chen G, et al. Role of osteopontin in synovial Th17 differentiation in rheumatoid arthritis. Arthr Rheum. 2010;62(10):2900–8.

    Article  CAS  Google Scholar 

  187. Gao SG, et al. Elevated osteopontin level of synovial fluid and articular cartilage is associated with disease severity in knee osteoarthritis patients. Osteoarthr Cartil. 2010;18(1):82–7.

    Article  CAS  Google Scholar 

  188. Gattorno M, et al. Synovial expression of osteopontin correlates with angiogenesis in juvenile idiopathic arthritis. Rheumatology (Oxford). 2004;43(9):1091–6.

    Article  CAS  Google Scholar 

  189. Han Z, et al. Relationship between miRNA-433 and SPP1 in the presence of fracture and traumatic brain injury. Exp Ther Med. 2021;22(3):928–928.

    Article  CAS  Google Scholar 

  190. Wosczyna MN, et al. Multipotent progenitors resident in the skeletal muscle interstitium exhibit robust BMP-dependent osteogenic activity and mediate heterotopic ossification. J Bone Miner Res. 2012;27(5):1004–17.

    Article  CAS  Google Scholar 

  191. Zhu Y, et al. MiR-19b-3p regulates osteogenic differentiation of PDGFRα(+) muscle cells by specifically targeting PTEN. Cell Biol Int. 2019;43(5):565–73.

    Article  CAS  Google Scholar 

  192. Huang J, et al. The role of COX-2 in mediating the effect of PTEN on BMP9 induced osteogenic differentiation in mouse embryonic fibroblasts. Biomaterials. 2014;35(36):9649–59.

    Article  CAS  Google Scholar 

  193. Zhao C, et al. miR-214 promotes osteoclastogenesis by targeting Pten/PI3k/Akt pathway. RNA Biol. 2015;12(3):343–53.

    Article  Google Scholar 

  194. Zheng H, et al. MicroRNA-181a/b-1 over-expression enhances osteogenesis by modulating PTEN/PI3K/AKT signaling and mitochondrial metabolism. Bone. 2019;123:92–102.

    Article  CAS  Google Scholar 

  195. Qin X, et al. miR-17-5p regulates heterotopic ossification by targeting ANKH in ankylosing spondylitis. Mol Ther Nucl Acids. 2019;18:696–707.

    Article  CAS  Google Scholar 

  196. Song H, et al. ACVR1, a therapeutic target of fibrodysplasia ossificans progressiva, is negatively regulated by miR-148a. Int J Mol Sci. 2012;13(2):2063–77.

    Article  CAS  Google Scholar 

  197. Arfat Y, et al. miR-208a-3p suppresses osteoblast differentiation and inhibits bone formation by targeting ACVR1. Mol Ther Nucl Acids. 2018;11:323–36.

    Article  CAS  Google Scholar 

  198. Huang Y, et al. Long noncoding RNA H19 promotes osteoblast differentiation via TGF-β1/Smad3/HDAC signaling pathway by deriving miR-675. Stem Cells. 2015;33(12):3481–92.

    Article  CAS  Google Scholar 

  199. Jin C, et al. Long non-coding RNA MIAT knockdown promotes osteogenic differentiation of human adipose-derived stem cells. Cell Biol Int. 2017;41(1):33–41.

    Article  CAS  Google Scholar 

  200. Liu L, et al. BRD4 promotes heterotopic ossification through upregulation of LncRNA MANCR. Bone Jt Res. 2021;10(10):668–76.

    Article  Google Scholar 

  201. Tracy KM, et al. Mitotically-associated lncRNA (MANCR) affects genomic stability and cell division in aggressive breast cancer. Mol Cancer Res. 2018;16(4):587–98.

    Article  CAS  Google Scholar 

  202. Nagasawa M, et al. Long non-coding RNA MANCR is a target of BET bromodomain protein BRD4 and plays a critical role in cellular migration and invasion abilities of prostate cancer. Biochem Biophys Res Commun. 2020;526(1):128–34.

    Article  CAS  Google Scholar 

  203. Lin L, et al. Adenovirus-mediated transfer of siRNA against Runx2/Cbfa1 inhibits the formation of heterotopic ossification in animal model. Biochem Biophys Res Commun. 2006;349(2):564–72.

    Article  CAS  Google Scholar 

  204. Mishra S, et al. Delivery of siRNA silencing Runx2 using a multifunctional polymer-lipid nanoparticle inhibits osteogenesis in a cell culture model of heterotopic ossification. Integr Biol (Camb). 2012;4(12):1498–507.

    Article  CAS  Google Scholar 

  205. Xue T, et al. Non-virus-mediated transfer of siRNAs against Runx2 and Smad4 inhibit heterotopic ossification in rats. Gene Ther. 2010;17(3):370–9.

    Article  CAS  Google Scholar 

  206. Shrivats AR, Hollinger JO. The delivery and evaluation of RNAi therapeutics for heterotopic ossification pathologies. In: Vunjak-Novakovic G, Turksen K, editors. Biomimetics and stem cells methods and protocols. New York: Springer; 2014. p. 149–60.

    Google Scholar 

  207. Chakraborty C, et al. Therapeutic advances of miRNAs: a preclinical and clinical update. J Adv Res. 2021;28:127–38.

    Article  CAS  Google Scholar 

  208. Dasgupta I, Chatterjee A. Recent advances in miRNA delivery systems. Methods Protoc. 2021;4(1):10.

    Article  CAS  Google Scholar 

  209. Elmén J, et al. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucl Acids Res. 2008;36(4):1153–62.

    Article  Google Scholar 

  210. Hong DS et al. MRX34, a liposomal miR-34 mimic, in patients with advanced solid tumors: final dose-escalation results from a first-in-human phase I trial of microRNA therapy. 2016, American Society of Clinical Oncology.

  211. van Zandwijk N, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96.

    Article  Google Scholar 

  212. Hueso M, et al. ncRNAs in therapeutics: challenges and limitations in nucleic acid-based drug delivery. Int J Mol Sci. 2021;22(21):11596.

    Article  CAS  Google Scholar 

  213. Huang C-K, Kafert-Kasting S, Thum T. Preclinical and clinical development of noncoding RNA therapeutics for cardiovascular disease. Circ Res. 2020;126(5):663–78.

    Article  CAS  Google Scholar 

  214. Dragomir M, Chen B, Calin GA. Exosomal lncRNAs as new players in cell-to-cell communication. Transl Cancer Res. 2017;7(Suppl 2):S243–52.

    Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

ŁP contributed to conceptualization; BM, IG, EB, ŁP, and AS contributed to writing—original draft preparation; BM, IG, EB, MAC, PŁ, PŁ, and AS contributed to writing, review, and editing; BM visualized the study; EB and ŁP supervised the study. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Łukasz Pulik.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pulik, Ł., Mierzejewski, B., Sibilska, A. et al. The role of miRNA and lncRNA in heterotopic ossification pathogenesis. Stem Cell Res Ther 13, 523 (2022). https://doi.org/10.1186/s13287-022-03213-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-022-03213-3

Keywords