Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 18.
Published in final edited form as: Semin Immunopathol. 2019 Oct 7;41(5):573–582. doi: 10.1007/s00281-019-00761-4

Osteoclastic microRNAs and their translational potential in skeletal diseases

Kazuki Inoue 1,2, Shinichi Nakano 1, Baohong Zhao 1,2,3
PMCID: PMC7027942  NIHMSID: NIHMS1557405  PMID: 31591677

Abstract

Skeleton undergoes constant remodeling process to maintain healthy bone mass. However, in pathological conditions, bone remodeling is deregulated, resulting in unbalanced bone resorption and formation. Abnormal osteoclast formation and activation play a key role in osteolysis, such as in rheumatoid arthritis and osteoporosis. As potential therapeutic targets or biomarkers, miRNAs have gained rapidly growing research and clinical attention. miRNA-based therapeutics is recently entering a new era for disease treatment. Such progress is emerging in treatment of skeletal diseases. In this review, we discuss miRNA biogenesis, advances in the strategies for miRNA target identification, important miRNAs involved in osteoclastogenesis and disease models, their regulated mechanisms, and translational potential and challenges in bone homeostasis and related diseases.

Keywords: Osteoclast, microRNA, Rheumatoid arthritis

Introduction

Bone tissue is a highly dynamic organ that undergoes constant remodeling process to maintain healthy bone mass. Bone remodeling requires a delicate balance between bone resorption mediated by osteoclasts and new bone formation mediated by osteoblasts/osteocytes. Approximately 5–10% of adult skeleton is remodeled annually, which differs greatly from other tissues in the body and makes bone a unique organ. Normal bone remodeling provides a necessary mechanism for adapting the skeleton to changing biomechanical influences and repairing bone damage. However, in pathological conditions, bone remodeling is deregulated, which results in unbalanced bone resorption and formation. Many disease settings, including rheumatoid arthritis (RA), psoriatic arthritis, peri-odontitis, peri-prosthetic loosening, and osteoporosis, are associated with bone destruction [1, 2]. Bone loss (osteolysis) is a major cause of morbidity and disability in these patients and significantly reduces their quality of life (QOL) and increases risk of mortality.

Giant multinucleated osteoclasts, derived from monocyte/macrophage lineage, are exclusively responsible for bone resorption. In pathological conditions, abnormal osteoclast formation and activation play a pivotal role in osteolysis. Comprehensive understanding of the osteoclastic regulatory mechanisms in both homeostatic and disease settings will facilitate development of novel or complementary therapeutic strategies to benefit patients for suppressing pathologic bone resorption. A great amount of work has focused on the effects of protein-coding genes on osteoclastogenesis. Studies from the last decade demonstrate the importance of microRNAs (miRNAs) in various biological and pathological settings, such as cell differentiation, proliferation, apoptosis, cancers, inflammatory disorders, metabolic diseases, and neurological diseases [35]. As potential therapeutic targets or biomarkers, miRNAs have recently gained rapidly growing research and clinical attention [612]. Targeting miRNAs has shown promising therapeutic potential and efficacy in several clinical trials, such as in the treatment of cancer, diabetes, and hepatitis C [1315]. This inspiring development highlights the clinical significance of a new era of miRNA-based therapeutics. In this review, we discuss miRNA biogenesis, advances in the strategies for miRNA target identification, important miRNAs involved in osteoclastogenesis and disease models, their regulated mechanisms, and translational potential and challenges in bone homeostasis and related diseases.

miRNA biogenesis

MicroRNAs (miRNAs) are small (~ 22 nucleotides), single-stranded non-coding RNAs that suppress the expression of their target mRNAs via post-transcriptional regulation [6, 16, 17]. miRNAs bind to partially complementary sequences in their target mRNAs but with perfect base pairing between the miRNA “seed region” (sequence of nucleotides 2–7 at the 5′ end of the miRNA) and the targeted sequences (microRNA response elements (MREs)) in 3′ untranslated region (3′-UTR) of mRNAs [18]. miRNAs repress gene expression by degradation or translational inhibition of specific target mRNAs, or a combination of these mechanisms [19]. Mature miRNAs are generated from long precursor RNAs, called primary miRNAs (pri-miRNAs), which are transcribed by RNA polymerase II [20]. In the nucleus, long pri-miRNAs are cropped by the microprocessor complex, which is consisted of the RNase III enzyme Drosha and RNA binding protein DiGeorge syndrome critical region 8 (DGCR8). This process produces hairpin-shaped pre-miRNAs (~ 60–70 nucleotides) [21, 22]. Pre-miRNAs are then exported to the cytoplasm by exportin 5 (XPO5) and further cleaved by the RNase III enzyme DICER [23]. DICER cleaves the double-stranded RNA stem and the terminal loop sequence of pre-miRNAs, and then a mature ~ 22 nucleotide miRNA/miRNA* duplex, containing a guide and a passenger strand (passenger strand designated with asterisk), is formed [24]. Subsequently, the guide strand of the mature miRNA is incorporated into miRNA-containing RNA-induced silencing complex (miRISC), which comprises DICER, transactivating response RNA-binding protein (TRBP), and Argonaute proteins (AGO1–4) [25, 26]. miRISC is guided by the guide strand to complementary target mRNAs and induces mRNA degradation, translational repression, or a combination of the two mechanisms. Each miRNA can repress multiple target mRNAs. Those target mRNAs sharing the same MREs may compete for miRNA binding and thus affect the expression of each other.

Advances in the strategies for miRNA target identification

Individual miRNAs often target multiple mRNA targets to regulate various biological processes. Identification of miRNA targets can reveal molecular mechanisms and regulatory networks of each miRNA. Recently, several bioinformatics tools, such as TargetScan, miRanda, PicTar, and DIANA-microT, have been developed to predict miRNA targets [2730]. These bioinformatics prediction algorithms are based on seed-pairing rules, the secondary structure of the 3′ UTR and evolutionary conservation. Traditional seed pairing rules predict miRNA-mRNA target recognition by a perfect complimentary sequence pairing between the miRNA “seed region” and the 3′-UTR of mRNAs [31]. The conventional ways identifying target mRNAs include (1) screening putative targets of each miRNA by bioinformatics tools, (2) validating the expression of target genes by qPCR and western blotting in combination with overexpression and/or inhibition of the miRNA of interest to examine whether there is an inverse correlation, and (3) a reporter assay testing the activity of the 3′ UTR of the predicted target genes (wild-type or mutated miRNA-pairing sequences) to corroborate a direct targeting by the miRNA. These bioinformatics tools, however, typically predict hundreds of targets for each miRNA, leading to a laborious and time-consuming way to identify authentic targets that have impacts on a biological process of interest.

Recent advances in high-throughput sequencing technology have developed several experimental approaches to identify endogenous miRNA targets on a genome-wide scale. Combination of the genome-wide profile of miRNA expression by miRNA-seq and genome-wide changes of mRNA expression levels by mRNA-seq following miRNA overexpression/inhibition significantly improve the efficiency and accuracy of miRNA target identification [32, 33]. In addition, tagged miRNA pull-down approach has successfully identified target mRNAs [34]. In this method, biotin-tagged microRNA mimics are introduced into cells and the biotinylated microRNA/mRNA complexes are purified by streptavidin-coated beads, then the captured mRNAs are analyzed by high-throughput sequencing. This approach is powerful yet not based on physiological conditions because of transfection of miRNA mimics, which needs attention when using this approach. On the other hand, several genome-wide approaches are developed based on immunoprecipitation of miRISC proteins, such as RIP-Seq (ribonucleoprotein immunoprecipitation followed by high-throughput sequencing) [35], HITS-CLIP (high-throughput sequencing of RNAs isolated by crosslinking immunoprecipitation) [36], PAR-CLIP (photoactivatable-ribonucleoside-enhanced crosslinking and immunoprecipitation) [37], and CLASH (crosslinking, ligation, and sequencing of hybrids) [38]. These techniques take advantage of crosslinking of endogenous miRNAs/mRNAs and proteins followed by immunoprecipitation with antibodies against miRISC proteins. Subsequently, fragmentation of the pull-downed RNAs with RNase followed by high-throughput sequencing is performed to identify miRNA-binding sites. Furthermore, some groups have focused on studying translational inhibition by miRNA and developed a ribosome profiling approach using miRNA mimics to identify targets [39]. Because a high density of ribosomes usually binds to translationally active mRNAs, high-throughput sequencing of ribosome-bound mRNAs in the presence or absence of miRNA mimics is able to identify target mRNAs.

These advances in high-throughput sequencing technologies and computational approaches have enabled significant progress in the discovery of miRNA targets on a genome-wide scale. Biological function of these miRNA-target interactions needs further exploration in diverse settings. Nonetheless, these high-throughput sequencing strategies greatly benefit miRNA target identification.

Most of early studies used in vitro osteoclast cultures, while recent studies often take advantage of both in vitro and in vivo systems to investigate miRNA functions. Due to space limitation, we will discuss a few miRNAs that play important roles in osteoclast differentiation, fusion, resorbing activity, survival, osteoclast-to-osteoblast communication, and diseases associated with bone destruction.

miR-21

miR-21 is upregulated by receptor activator of nuclear factor-kappaB ligand (RANKL) and promotes osteoclastogenesis through targeting programmed cell death 4 (PDCD4) protein levels, which subsequently regulate the c-Fos-NFATc1 axis [40]. A later study showed that estrogen suppresses miR-21 biogenesis, which increases FasL protein levels because FasL is a target of miR-21. The enhanced autocrine FasL binds to Fas and induces apoptosis in osteoclasts. Thus, miR-21 also controls osteoclast survival in response to estrogen [41].

miR-31

miR-31, induced by RANKL, is a positive regulator for cyto-skeleton organization and bone resorptive activity of osteoclasts by targeting RhoA [42].

miR-155

miR155 is a well-studied miRNA that plays crucial roles in various immune cells in both innate and adaptive immunity [4348]. In monocyte/macrophage lineage, miR155 is often induced by inflammatory stimuli, such as LPS and TNF, and regulates proliferation, differentiation, and function of macrophages and dendritic cells [43, 44, 49, 50]. In contrast to the expression pattern and activating function in macrophages, miR-155 is downregulated by RANKL signaling and impairs RANKL-induced osteoclast differentiation by targeting microphthamia-associated transcription factor (MITF) and PU.1. Interestingly, IFNβ induces miR-155, which represses osteoclast differentiation by targeting suppressor of cytokine signaling 1 (SOCS1) and MITF. Taken together, miR-155 acts as an inhibitory miRNA in osteoclastogenesis [51].

miR-223

miR-223 is first identified as a myeloid regulator, potentially controlled by the transcription factor PU.1 [52]. The expression of miR-223 is strongly upregulated in myeloid cells, including neutrophils and macrophages, and it is an important regulator of myeloid cell differentiation. Notably, miR-223 expression is elevated in RA patients [53, 54], and is overexpressed in CD68+ macrophages, CD14+ monocytes, and CD4+ T cells isolated from the synovium of RA patients [55]. miR-223 is expressed in osteoclast precursors and negatively regulates osteoclastogenesis by targeting nuclear factor I A (NFI-A) [56], which is required for upregulating M-CSF receptor levels that in turn induces the expression of PU.1 and c-Fos [57].

miR-7b

The immune function of miR-7 in autoreactive B cells from systemic lupus erythematosus (SLE) patients was reported by Wu et al. [58]. B cell hyperresponsiveness in SLE is caused by enhanced B cell receptor (BCR) signaling, which is mediated by the Pten/phosphatidylinositol 3-kinase (PI3K) pathway [59]. Wu et al. revealed that Pten expression is decreased in B cells from SLE patients and inversely correlated with disease activity. miR-7 expression is upregulated in the SLE B cells and targeting Pten by miR-7 contributes to B cell hyperresponsiveness in SLE.

Multinucleation formed by the cell-cell fusion of mononuclear osteoclast precursors is an important step for osteoclast maturation. Dendritic cell-specific transmembrane protein (DC-STAMP) is a key regulator of osteoclast precursor (OCP) fusion [60]. DC-STAMP expression is positively regulated by NFATc1, c-Fos, and strawberry notch homolog 2 (Sbno2) [61]. However, the post-transcriptional regulation of DC-STAMP expression is largely unclear. miR-7b is identified as a negative regulator of osteoclastogenesis and cell-cell fusion by directly targeting DC-STAMP [62]. Overexpression of miR-7b represses other fusogenic genes (CD47, ATP6v0d2 and OC-STAMP) as well as osteoclast-specific genes (Nfatc1 and OSCAR) via DC-STAMP inhibition [62].

miR-34a

miR-34a is known to modulate macrophage differentiation and functions. miR-34a is highly expressed in alveolar macrophages and mediates efferocytosis by targeting Axl, a receptor tyrosine kinase-recognizing apoptotic cells, and silent information regulator T1 (Sirt1) [63]. Another group reported the function of miR-34a in pre-B cell-to-macrophage transdifferentiation, in which miR-34a as a direct target of CCAAT/enhancer-binding protein-α (C/EBPα), together with miR223, inhibits Lef1 expression to achieve C/EBPα-mediated silencing of the B cell-specific gene program and transdifferentiation into functional macrophages [64, 65].

Pathological osteoclast differentiation contributes to both osteoporosis and osteolytic bone metastases of cancer. Krzeszinski et al. identified miR-34a as a novel negative regulator of osteoclastogenesis, bone resorption, and the bone metastatic niche [66]. miR-34a expression level is decreased during osteoclast differentiation. Osteoclastogenesis from both mouse bone marrow-derived macrophages (BMMs), and human peripheral blood mononuclear cells is suppressed by miR-34a overexpression but promoted by miR-34a inhibition. Osteoclastic miR-34a-overexpressing transgenic mice exhibit reduced bone resorption and elevated bone mass. On the other hand, miR-34a knockout mice show a complementary bone phenotype. Under pathological conditions, such as ovariectomy (OVX)-induced osteoporosis and bone metastasis of breast or skin cancers, osteoclastic miR-34a overexpression impedes pathological bone resorption and bone metastasis. Moreover, treatment with miR-34a encapsulated in chitosan-nanoparticles effectively attenuates both osteoporosis and cancer bone metastases. Mechanistically, transforming growth factor-β-induced factor 2 (Tgif2) is identified as a direct target of miR-34a in these settings.Tgif2 knockout mice exhibit reduced bone resorption and higher bone mass. Tgif2 deletion abolishes the anti-osteoclastogenic effects of miR-34a. Taken together, miR-34a is a key suppressor of osteoclastogenesis and augmentation of miR-34a activity has a strong therapeutic potential for pathological bone resorption associated with osteoporosis and cancer bone metastases.

miR-124

miR-124 was originally identified as a key regulator that controls the immune function of microglia, tissue-resident macrophages in the central nervous system (CNS), by directly suppressing the transcription factor CCAAT/enhancer-binding protein-α (C/EBP-α) and its downstream target PU.1 [67]. miR-124 expression is decreased during osteoclastogenesis. miR-124 suppresses osteoclastogenesis of mouse BMMs by inhibiting the protein expression of Nfatc1, the master transcription factor for osteoclast differentiation [68]. Furthermore, Nakamachi et al. [69] reported that miR-124 is downregulated in the ankles of adjuvant-induced arthritis (AIA) rats. Injection of pre-miR-124 into the ankles of AIA rats strongly ameliorates bone destruction with attenuation of synoviocyte proliferation, leukocyte infiltration into synovial tissue and the destruction of cartilage. Osteoclastogenesis in the joints of AIA rats is also suppressed with pre-miR-124 treatment via directly targeting Nfatc1.

miR-141

miR-141 is as an important regulator involved in osteolytic bone metastasis, which occurs frequently in late stage of breast and bladder cancers, often leading to pathological bone fractures. By genome-wide screening of miRNAs in osteoclasts in response to conditioned medium (CM) obtained from the culture of bone metastatic tumors, such as 4T1.2 and TSU-Pr-B2 cells, Ell et al. found that miR-141 was downregulated by the CM treatment [70]. Ectopic expression of miR-141 inhibits osteoclast differentiation through inhibiting the expression of Mitf and Calcr (calcitonin receptor). Systemic treatment of miR-141 inhibits in vivo osteoclast differentiation and increases trabecular bone. miR-141 treatment in a mouse model of breast cancer bone metastasis also downregulates in vivo osteoclastogenesis and suppresses bone metastasis [70]. Recently, Yang and colleagues showed the importance of miR-141 in osteoclast differentiation and bone resorption in aged rhesus monkeys [71]. They found that the expression levels of miR-141 were downregulated in aged osteoporotic patients and aged rhesus monkeys. Selective delivery of miR-141 into the aged rhesus monkeys using Asp (Aspartic acid) 8-PU (polyurethane) nanoparticles, which specifically target bone-resorption surfaces, inhibited in vivo osteoclast differentiation and increased trabecular bone mass. Mechanistically, miR-141 inhibits osteoclast differentiation by targeting Calcr and EphA2 (ephrin type-A receptor 2 precursor). Collectively, these studies indicate an important role for miR-141 in suppressing bone resorption in primates and provide experimental evidence for future clinical application of miRNAs in treatment for osteolytic bone metastasis and osteoporosis.

miR-503

miR-503 was first identified as one of the tumor-associated miRNAs inducing dendritic cell apoptosis by targeting Bcl2 [72]. Later, Chen et al. [73] performed miRNA microarray to compare the miRNA expression profiles in CD14+ peripheral blood mononuclear cells (PBMCs) from postmenopausal osteoporosis patients and postmenopausal healthy women. They found that miR-503 shows the most dramatic downregulation in CD14+ cells isolated from osteoporosis patients. RANKL-induced osteoclastogenesis is inhibited by overexpression of miR-503 in CD14+ PBMCs. Conversely, miR-503 inhibition in CD14+ PBMCs enhances osteoclastogenesis. miR-503 directly targets receptor activator of nuclear factor κ B (RANK), the receptor for RANKL. Specific agomir for miR-503 prevents OVX mice from significant bone loss. Thus, miR-503 may contribute to the pathogenesis of postmenopausal osteoporosis and could be a potential biomarker and therapeutic target for postmenopausal osteoporosis.

miR-214

Osteoclasts and osteoblasts control bone homeostasis by communicating with each other through coupling factors, such as RANKL, OPG, TGF-β1, IGF-1, Sema4D, Sema3A, EphrinB2, Cthrc1, and Wnt16 [7479]. Exosomes are emerging as essential messengers delivering bioactive molecules, such as mRNAs, microRNAs, and proteins, to mediate inter-cellular communication [80]. Recently, miR-214–3p was identified as an osteoclast-derived exosomal miRNA communicating with osteoblasts [81]. Osteoclastic miR-214–3p is increased with elevation of serum exosomal miR-214–3p in elderly women with fractures and in OVX mice [81]. Osteoclast-specific miR-214–3p overexpression mice (OC-miR-214–3p) exhibit increased serum exosomal miR-214–3p and reduced bone formation. Injection of antagomiR-214–3p enveloped by (D-Asp8)-liposome [82], which can target osteoclasts, rescues low-bone-mass phenotypes in OC-miR-214–3p mice. Moreover, in co-culture system, osteoclast-derived exosomal miR-214–3p is able to transfer to osteoblasts and inhibit osteoblast activity. Treatment with supernatant exosomes derived from the cultures of primary OC-miR-214–3p osteoclasts reduces bone formation in vivo, whereas osteoclast-targeted antagomiR-214–3p treatment enhances bone formation in aged OVX mice. The exosomal miR-214–3p derived from osteoclasts inhibits osteoblast activity via inhibiting the protein expression of an important osteogenic transcription factor ATF4, a target of miR-214–3p in osteoblasts [81, 83, 84]. In addition, miR-214–3p expression is increased during osteoclastogenesis and plays a positive role in osteoclast differentiation and activity by directly targeting phosphatase and tensin homolog (Pten) and regulating PI3K/Akt pathway [85]. Previous studies show that miR-214–3p also targets Pten to enhance T cell activation and proliferation [86, 87]. Osteoclast-specific miR-214 transgenic mice exhibit increased osteoclastogenesis and resorption activity with reduced Pten protein levels and reduced bone mass. Upregulated miR-214–3p expression is also observed in bone specimens from breast cancer patients with osteolytic bone metastasis [88]. The increase of miR-214–3p is correlated with the elevation of bone resorption during the development of osteolytic bone metastasis in nude mice xenografted with human breast cancer MDA-MB-231 cells. Osteoclast-specific miR-214–3p-deficient nude mice show resistance to osteolytic bone metastasis. In this study, TNF receptor-associated factor 3 (Traf3) is identified as a direct target of miR-214–3p, and the increased bone resorption observed in OC-miR-214–3p mice is attenuated with administration of Traf3 3′UTR-containing plasmid encapsulated with (D-Asp8)-liposome [88]. Delivery of osteoclast-targeted antagomiR-214–3p recovers TRAF3 protein levels and attenuates bone resorption and osteolytic bone metastasis. Taken together, these studies strongly indicate the therapeutic benefits of targeting miR-214–3p through regulating both osteoclasts and osteoblasts for patients with abnormal bone remodeling especially in cancer bone metastasis.

miR-182/miR-183

miR-183 cluster is comprised of miR-182, miR-183, and miR-96 [89]. miR-96 is undetectable in osteoclastogenesis [32]. miR-183 expression is elevated by RANKL and positively regulates RANKL-induced osteoclastogenesis via heme oxygenase-1 (HO-1) suppression in in vitro osteoclastogenesis [90]. The important function of miR-182 in cell growth, cell fate decision, cancer, T lymphocyte expansion, and Th17 function was just recently appreciated [91, 92]. We initially performed high-throughput miRNA-sequencing and obtained a genome-wide profile of miRNA expression induced by TNFα in mouse BMMs [32]. Based on this database, we identified that miR-182 is a new regulator in TNF-induced inflammatory osteoclast differentiation in vitro [32]. We then elucidated the role of miR-182 in vivo in physiological bone metabolism and pathological conditions, such as those that occur in osteoporosis and inflammatory arthritis [93]. Pathologic bone destruction is a severe consequence and characteristic of diseases such as rheumatoid arthritis (RA) and postmenopausal osteoporosis, in which osteoclasts are directly responsible for osteolysis. miRNA-based therapeutics is recently entering a new era for disease treatment; however, such progress is quite underdeveloped in treatment of skeletal diseases. We identified miR-182 as a key osteoclastogenic regulator, provided strong translational implications of targeting miR-182 in pathologic bone destruction, uncovered a novel miRNA-orchestrated regulatory network that controls interferon pathway in skeleton, and revealed significant correlation between miR-182 and human RA disease.

The principal findings of our study on miR-182 and their significance include (1) strong translational implications: using complementary gain and loss-of-function approaches (myeloid-specific miR-182 KO and Tg mice), we identify miR-182 as a key positive regulator of osteoclastogenesis, bone homeostasis, and pathologic bone destruction. To investigate translational significance of targeting miR-182, we applied two disease models in our study: ovariectomy (OVX)-induced osteoporosis that mimics postmenopausal osteoporosis and inflammatory arthritis that mimics RA. Inhibition of miR-182 by genetic ablation or pharmacological inhibitors completely counteracts bone loss in both disease models, indicating a robust bone protection effect by miR-182 inhibition. Of note, this protection is not attributed to the basal bone mass level in the miR-182-deficient mice. To increase the stability, efficacy, and specificity of cellular targeting of the miR-182 inhibitors, we utilized chitosan nanoparticles as delivery vehicles. The chitosan nanoparticles used were packaged with a specific formula that enables them to have the highest bio-distribution in bone marrow and target myeloid osteoclast cell linage. Importantly, the nanoparticles per se are safe and do not affect bone mass. Inhibition of miR-182 does not show undesired side effects, such as immune suppression in these disease models. Thus, our findings highlight promising therapeutic implications of miR-182 inhibition in these diseases and provide proof-of-principle that targeting miR-182 may have clinical utility to treat bone loss. (2) Novel molecular mechanisms: we identify PKR (protein kinase double-stranded RNA-dependent) as a new and essential miR-182 target that is a novel inhibitor of osteoclastogenesis. PKR attenuates osteoclast differentiation via regulation of endogenous IFN-β-mediated autocrine feedback loop. Osteoclastogenesis is determined by the balance between osteoclastogenic and anti-osteoclastogenic factors. Although previously unclear how IFN-β mediated inhibitory loop was downregulated, our studies, for the first time, uncovered an important mechanism that miR-182-PKR axis is responsible for suppressing autocrine IFN-β signaling. These findings indicate a conceptually new model, in which a previously unrecognized regulatory circuit, orchestrated by miR-182-PKRIFN-β axis, fine tunes the osteoclastogenic network. (3) Significant disease correlation: for the first time, our results reveal significant changes of the miR-182-PKR-IFN-β axis with higher miR-182 levels, and lower PKR and IFN-β levels in the PBMCs isolated from RA patients than from healthy donors. Serum TNF levels affect the expression of this axis, and TNFi therapy (Enbrel) reverses the expression of miR-182, PKR, and IFN-β towards healthy donors’ levels. Importantly, the osteoclastogenic capacity of RA PBMCs is strongly correlated with the expression levels of miR-182 (positively), PKR (negatively), and IFN-β (negatively). Functional analysis of miR-182 in human PBMCs by gene silencing reveals its key osteoclastogenic role. Thus, as evidenced from our murine and human data, both the regulatory pattern of the miR-182-PKR-IFN-β axis and the miR-182 function are well conserved. These human RA data therefore further support a translational promise of targeting miR-182 in diseases associated with bone loss, such as RA.

Perspectives and challenges of miRNA-based therapeutics

miRNAs exert functions through their specific targets and the downstream pathways mediated by the targets. By binding to complementary “seed” region in target mRNAs, different miRNAs target different genes. However, the target genes regulated by the same miRNA are often variable depending on cell and tissue types. Even in the same cell type, the same miRNA targets can be different in response to distinct stresses or disease settings, presumably due to diverse gene expression and regulation profiles in different conditions. Thus, miRNA-targeted gene regulation is highly specific and quite sensitive to environmental changes. In addition, the same biological process, for example osteoclast differentiation, can be regulated by multiple miRNAs, whose functions may or may not compensate each other in this process. It is therefore important to take into consideration of potential side effects from variable targets by a specific miRNA targeting in preclinical development and clinical trials. Recent studies have shown that mRNA, transcribed pseudogenes, long non-coding RNAs (lncRNA), and circular RNAs (circRNA) sequester miRNAs and block them from binding to their mRNA targets. Those RNAs are referred to as competing endogenous RNAs (ceRNAs) [94, 95], functioning as molecular sponges for miRNAs and de-repress all target genes of a specific miRNA. These findings add another layer of complexity of miRNA-mediated gene regulation. Therefore, understanding of not only miRNA-target mRNA interactions but also miRNA-ceRNA networks will help fully delineate the mechanisms of miRNA-mediated gene regulation.

Despite a significant progress in miRNA therapeutics, only a small number of miRNA mimics or inhibitors have entered clinical development. Another challenge is the design of miRNA delivery approaches that can ideally make the miRNA-based drugs stable and enable tissue-specific targeting, meanwhile minimizing potential toxicities and off-target effects. Naked small RNA molecules are easily degradable. Chemical modification of the nucleotide back-bone of miRNA mimics or inhibitors, such as modification with locked nucleic acid (LNA), have improved their binding affinity and stability. For example, in our studies, the miR-182 inhibitor has LNA modification. The initial preliminary results however showed that a large amount of the miR-182 inhibitors (~ 1 mg daily) was required to suppress osteoclastogenesis in vivo, indicating a delivery vehicle is necessary to reduce amount. Indeed, recent in vivo delivery technologies, including nanoparticle systems, have enabled the first generation of miRNA-based agents to move into the pre-clinical development and clinic trials. Chitosan is a cationic polymer derived from chitin and has been extensively used for small RNA delivery in preclinical studies due to its biodegradability and low cellular toxicity. We applied chitosan nanoparticles to incorporate miR-182 inhibitors and reduced approximately 600 times of the amount of miR-182 inhibitors down to 5 μg every 3 days to suppress osteoclastogenesis. The low amount of miR-182 inhibitors using chitosan delivery system not only functions efficiently but also could lower off-targeting effects and cellular toxicity. Chitosan is FDA-approved safe for wound dressing and dietary use. Several animal toxicity studies, including our results, reported good safety in vivo [93, 96]. The nanoparticle formula decides the particle size and weight that usually delicately determine the specificity of targeting certain cells. For example, the chitosan formula can be optimized to facilitate targeting monocytes/macrophages and bone marrow [66, 93]. Other groups were successful by using aptamer-functionalized lipid nanoparticles, such as (D-Asp8)-liposome, or Asp (Aspartic acid) 8-PU (polyurethane) nanoparticles that specifically target bone-resorption surfaces, as delivery vehicles in animal models to target osteoclasts [71, 82, 88]. Nonetheless, in order to eventually achieve a successful clinical application, precise identification of miRNA targets in different diseases and development of more osteoclast-specific targeting delivery approaches should be conducted when developing therapeutic applications to treat osteolysis.

The challenges, as described above, give rise to new opportunities for miRNA-based therapeutics. As reviewed in this article, recent miRNA studies provide a proof of concept for the efficacy of therapeutic targeting of miRNAs to prevent or treat bone loss based on the genetic evidence from both in vitro and in vivo systems, correlation between miRNA expression levels and osteoclastogenic capacity in skeletal diseases, and the in vivo pharmacological results obtained from various animal disease models. These promising studies highlight the translational implications of miRNA-based therapeutics in treating osteolytic diseases, especially the refractory bone resorption such as that occurs in RA, or life-threatening bone destruction associated with cancer bone metastasis.

Acknowledgments

We thank Courtney Ng for the critical review of this manuscript.

Funding information This work was supported by grants from the National Institutes of Health (NIH R01 AR068970 and R01 AR071463 to B.Z.).

Footnotes

Conflict of interest The authors declare no conflict of interests.

Publisher's Disclaimer: Disclaimer The content of this manuscript is solely the responsibilities of the authors and does not necessarily represent the official views of the NIH.

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

References

  • 1.Schett G, Gravallese E (2012) Bone erosion in rheumatoid arthritis: mechanisms, diagnosis and treatment. Nat Rev Rheumatol 8(11): 656–664 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Goldring SR, Purdue PE, Crotti TN, Shen Z, Flannery MR, Binder NB, Ross FP, McHugh KP (2013) Bone remodelling in inflammatory arthritis. Ann Rheum Dis 72(Suppl 2):ii52–ii55 [DOI] [PubMed] [Google Scholar]
  • 3.Alvarez-Garcia I, Miska EA (2005) MicroRNA functions in animal development and human disease. Development. 132(21):4653–4662 [DOI] [PubMed] [Google Scholar]
  • 4.Ardekani AM, Naeini MM (2010) The role of microRNAs in human diseases. Avicenna J Med Biotechnol 2(4):161–179 [PMC free article] [PubMed] [Google Scholar]
  • 5.Meydan C, Shenhar-Tsarfaty S, Soreq H (2016) MicroRNA regulators of anxiety and metabolic disorders. Trends Mol Med 22(9): 798–812 [DOI] [PubMed] [Google Scholar]
  • 6.Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 116(2):281–297 [DOI] [PubMed] [Google Scholar]
  • 7.Hayes J, Peruzzi PP, Lawler S (2014) MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol Med 20(8):460–469 [DOI] [PubMed] [Google Scholar]
  • 8.He L, Hannon GJ (2004) MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 5(7):522–531 [DOI] [PubMed] [Google Scholar]
  • 9.Olive V, Minella AC, He L (2015) Outside the coding genome, mammalian microRNAs confer structural and functional complexity. Sci Signal 8(368):re2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Singh RP, Massachi I, Manickavel S, Singh S, Rao NP, Hasan S, Mc Curdy DK, Sharma S, Wong D, Hahn BH, Rehimi H (2013) The role of miRNA in inflammation and autoimmunity. Autoimmun Rev 12(12):1160–1165 [DOI] [PubMed] [Google Scholar]
  • 11.Srinivasan S, Selvan ST, Archunan G, Gulyas B, Padmanabhan P (2013) MicroRNAs -the next generation therapeutic targets in human diseases. Theranostics. 3(12):930–942 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Stanczyk J, Pedrioli DM, Brentano F, Sanchez-Pernaute O, Kolling C, Gay RE, Detmar M, Gay S, Kyburz D (2008) Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum 58(4):1001–1009 [DOI] [PubMed] [Google Scholar]
  • 13.Rupaimoole R, Slack FJ (2017) MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov 16(3):203–222 [DOI] [PubMed] [Google Scholar]
  • 14.Chakraborty C, Sharma AR, Sharma G, Doss CGP, Lee SS (2017) Therapeutic miRNA and siRNA: moving from bench to clinic as next generation medicine. Mol Ther Nucleic Acids 8:132–143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y et al. (2013) Treatment of HCV infection by targeting microRNA. N Engl J Med 368(18):1685–1694 [DOI] [PubMed] [Google Scholar]
  • 16.Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T (2001) Identification of novel genes coding for small expressed RNAs. Science. 294(5543):853–858 [DOI] [PubMed] [Google Scholar]
  • 17.AJ S, S L, JJ S (2008) et al. MIcrorna expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. Jama. 299:425–436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bartel DP (2009) MicroRNAs: target recognition and regulatory functions. Cell. 136(2):215–233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ling H, Fabbri M, Calin GA (2013) MicroRNAs and other noncoding RNAs as targets for anticancer drug development. Nat Rev Drug Discov 12(11):847–865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN (2004) MicroRNA genes are transcribed by RNA polymerase II. EMBO J 23(20):4051–4060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R (2004) The microprocessor complex mediates the genesis of microRNAs. Nature. 432(7014):235–240 [DOI] [PubMed] [Google Scholar]
  • 22.Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S et al. (2003) The nuclear RNase III Drosha initiates microRNA processing. Nature. 425(6956):415–419 [DOI] [PubMed] [Google Scholar]
  • 23.Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U (2004) Nuclear export of microRNA precursors. Science. 303(5654):95–98 [DOI] [PubMed] [Google Scholar]
  • 24.Park JE, Heo I, Tian Y, Simanshu DK, Chang H, Jee D, Patel DJ, Kim VN (2011) Dicer recognizes the 5′ end of RNA for efficient and accurate processing. Nature. 475(7355):201–205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R (2005) TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 436(7051):740–744 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Peters L, Meister G (2007) Argonaute proteins: mediators of RNA silencing. Mol Cell 26(5):611–623 [DOI] [PubMed] [Google Scholar]
  • 27.Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB (2003) Prediction of mammalian microRNA targets. Cell. 115(7): 787–798 [DOI] [PubMed] [Google Scholar]
  • 28.John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS (2004) Human MicroRNA targets. PLoS Biol 2(11):e363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M et al. (2005) Combinatorial microRNA target predictions. Nat Genet 37(5):495–500 [DOI] [PubMed] [Google Scholar]
  • 30.Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T, Reczko M, Filippidis C, Dalamagas T, Hatzigeorgiou AG (2013) DIANA-microT web server v5.0: service integration into miRNA functional analysis workflows. Nucleic Acids Res 41(Web Server issue):W169–W173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Filipowicz W, Bhattacharyya SN, Sonenberg N (2008) Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 9(2):102–114 [DOI] [PubMed] [Google Scholar]
  • 32.Miller CH, Smith SM, Elguindy M, Zhang T, Xiang JZ, Hu X, Ivashkiv LB, Zhao B (2016) RBP-J-regulated miR-182 promotes TNF-alpha-induced osteoclastogenesis. J Immunol 196(12):4977–4986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Maeda Y, Farina NH, Matzelle MM, Fanning PJ, Lian JB, Gravallese EM (2016) Synovium-derived microRNAs regulate bone pathways in rheumatoid arthritis. J Bone Miner Res [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Krishnan K, Steptoe AL, Martin HC, Wani S, Nones K, Waddell N, Mariasegaram M, Simpson PT, Lakhani SR, Gabrielli B, Vlassov A, Cloonan N, Grimmond SM (2013) MicroRNA-182–5p targets a network of genes involved in DNA repair. RNA. 19(2):230–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhao J, Ohsumi TK, Kung JT, Ogawa Y, Grau DJ, Sarma K, Song JJ, Kingston RE, Borowsky M, Lee JT (2010) Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell 40(6):939–953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Licatalosi DD, Mele A, Fak JJ, Ule J, Kayikci M, Chi SW, Clark TA, Schweitzer AC, Blume JE, Wang X, Darnell JC, Darnell RB (2008) HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature. 456(7221):464–469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, Berninger P, Rothballer A, Ascano M Jr, Jungkamp AC, Munschauer M, Ulrich A, Wardle GS, Dewell S, Zavolan M, Tuschl T (2010) Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell. 141(1):129–141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Helwak A, Kudla G, Dudnakova T, Tollervey D (2013) Mapping the human miRNA interactome by CLASH reveals frequent non-canonical binding. Cell. 153(3):654–665 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Guo H, Ingolia NT, Weissman JS, Bartel DP (2010) Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466(7308):835–840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sugatani T, Vacher J, Hruska KA (2011) A microRNA expression signature of osteoclastogenesis. Blood. 117:3648–3657 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sugatani T, Hruska KA (2013) Down-regulation of miR-21 biogenesis by estrogen action contributes to osteoclastic apoptosis. J Cell Biochem 114:1217–1222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mizoguchi F, Murakami Y, Saito T, Miyasaka N, Kohsaka H (2013) miR-31 controls osteoclast formation and bone resorption by targeting RhoA. Arthritis Res Ther 15(5):R102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.O’Connell RM, Chaudhuri AA, Rao DS, Baltimore D (2009) Inositol phosphatase SHIP1 is a primary target of miR-155. Proc Natl Acad Sci U S A 106:7113–7118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D (2007) MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci U S A 104:1604–1609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Thai T-H, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y, Murphy A, Frendewey D, Valenzuela D, Kutok JL, Schmidt-Supprian M, Rajewsky N, Yancopoulos G, Rao A, Rajewsky K (2007) Regulation of the germinal center response by microRNA-155. Science. 316:604–608 [DOI] [PubMed] [Google Scholar]
  • 46.Vigorito E, Perks KL, Abreu-Goodger C, Bunting S, Xiang Z, Kohlhaas S, Das PP, Miska EA, Rodriguez A, Bradley A, Smith KGC, Rada C, Enright AJ, Toellner KM, MacLennan ICM, Turner M (2007) microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity. 27:847–859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Teng G, Hakimpour P, Landgraf P, Rice A, Tuschl T, Casellas R, Papavasiliou FN (2008) MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity. 28:621–629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Satoorian T, Li B, Tang X, Xiao J, Xing W, Shi W, Lau K-HW, Baylink DJ, Qin X (2016) MicroRNA223 promotes pathogenic T-cell development and autoimmune inflammation in central nervous system in mice. Immunology. 148:326–338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lind EF, Millar DG, Dissanayake D, Savage JC, Grimshaw NK, Kerr WG, Ohashi PS (2015) miR-155 upregulation in dendritic cells is sufficient to break tolerance in vivo by negatively regulating SHIP1. J Immunol 195(10):4632–4640 [DOI] [PubMed] [Google Scholar]
  • 50.Ceppi M, Pereira PM, Dunand-Sauthier I, Barras E, Reith W, Santos MA, Pierre P (2009) MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. Proc Natl Acad Sci U S A 106(8):2735–2740 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mann M, Barad O, Agami R, Geiger B, Hornstein E, Elaine Fuchs b miRNA-based mechanism for the commitment of multipotent progenitors to a single cellular fate departments of a molecular genetics and [DOI] [PMC free article] [PubMed]
  • 52.Fukao T, Fukuda Y, Kiga K, Sharif J, Hino K, Enomoto Y, Kawamura A, Nakamura K, Takeuchi T, Tanabe M (2007) An evolutionarily conserved mechanism for MicroRNA-223 expression revealed by microRNA gene profiling. Cell. 129:617–631 [DOI] [PubMed] [Google Scholar]
  • 53.Alevizos I, Illei GG (2010) MicroRNAs as biomarkers in rheumatic diseases. Nat Rev Rheumatol 6:391–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li YT, Chen SY, Wang CR, Liu MF, Lin CC, Jou IM, Shiau AL, Wu CL (2012) Amelioration of collagen-induced arthritis in mice by lentivirus-mediated silencing of microRNA-223. Arthritis Rheum 64:3240–3245 [DOI] [PubMed] [Google Scholar]
  • 55.Shibuya H, Nakasa T, Adachi N, Nagata Y, Ishikawa M, Deie M, Suzuki O, Ochi M (2013) Overexpression of microRNA-223 in rheumatoid arthritis synovium controls osteoclast differentiation. Mod Rheumatol 23:674–685 [DOI] [PubMed] [Google Scholar]
  • 56.Sugatani T, Hruska KA (2007) MicroRNA-223 is a key factor in osteoclast differentiation. J Cell Biochem 101:996–999 [DOI] [PubMed] [Google Scholar]
  • 57.Sugatani T, Hruska KA (2009) Impaired micro-RNA pathways diminish osteoclast differentiation and function. J Biol Chem 284: 4667–4678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wu XN, Ye YX, Niu JW, Li Y, Li X, You X, Chen H, Zhao LD, Zeng XF, Zhang FC, Tang FL, He W, Cao XT, Zhang X, Lipsky PE (2014) Defective PTEN regulation contributes to B cell hyperresponsiveness in systemic lupus erythematosus. Sci Transl Med 6(246):246ra99. [DOI] [PubMed] [Google Scholar]
  • 59.Okkenhaug K, Burger JA (2016) PI3K signaling in normal B cells and chronic lymphocytic leukemia (CLL). Curr Top Microbiol Immunol 393:123–142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yagi M, Miyamoto T, Sawatani Y, Iwamoto K, Hosogane N, Fujita N, Morita K, Ninomiya K, Suzuki T, Miyamoto K, Oike Y, Takeya M, Toyama Y, Suda T (2005) DC-STAMP is essential for cell-cell fusion in osteoclasts and foreign body giant cells. J Exp Med 202(3):345–351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Maruyama K, Uematsu S, Kondo T, Takeuchi O, Martino MM, Kawasaki T, Akira S (2013) Strawberry notch homologue 2 regulates osteoclast fusion by enhancing the expression of DC-STAMP. J Exp Med 210(10):1947–1960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dou C, Zhang C, Kang F, Yang X, Jiang H, Bai Y, Xiang J, Xu J, Dong S (2014) MiR-7b directly targets DC-STAMP causing suppression of NFATc1 and c-Fos signaling during osteoclast fusion and differentiation. Biochim Biophys Acta 1839(11):1084–1096 [DOI] [PubMed] [Google Scholar]
  • 63.McCubbrey AL, Nelson JD, Stolberg VR, Blakely PK, McCloskey L, Janssen WJ, Freeman CM, Curtis JL (2016) MicroRNA-34a negatively regulates efferocytosis by tissue macrophages in part via SIRT1. J Immunol 196(3):1366–1375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Xie H, Ye M, Feng R, Graf T (2004) Stepwise reprogramming of B cells into macrophages. Cell. 117(5):663–676 [DOI] [PubMed] [Google Scholar]
  • 65.Rodriguez-Ubreva J, Ciudad L, van Oevelen C, Parra M, Graf T, Ballestar E (2014) C/EBPa-mediated activation of microRNAs 34a and 223 inhibits Lef1 expression to achieve efficient reprogramming into macrophages. Mol Cell Biol 34(6):1145–1157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Krzeszinski JY, Wei W, Huynh H, Jin Z, Wang X, Chang TC, Xie XJ, He L, Mangala LS, Lopez-Berestein G, Sood AK, Mendell JT, Wan Y (2014) miR-34a blocks osteoporosis and bone metastasis by inhibiting osteoclastogenesis and Tgif2. Nature. 512(7515):431–435 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 67.Ponomarev ED, Veremeyko T, Barteneva N, Krichevsky AM, Weiner HL (2011) MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-alpha-PU.1 pathway. Nat Med 17(1):64–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lee Y, Kim HJ, Park CK, Kim YG, Lee HJ, Kim JY, Kim HH (2013) MicroRNA-124 regulates osteoclast differentiation. Bone. 56:383–389 [DOI] [PubMed] [Google Scholar]
  • 69.Nakamachi Y, Ohnuma K, Uto K, Noguchi Y, Saegusa J, Kawano S (2016) MicroRNA-124 inhibits the progression of adjuvant-induced arthritis in rats. Ann Rheum Dis 75(3):601–608 [DOI] [PubMed] [Google Scholar]
  • 70.Ell B, Mercatali L, Ibrahim T, Campbell N, Schwarzenbach H, Pantel K, Amadori D, Kang Y (2013) Tumor-induced osteoclast miRNA changes as regulators and biomarkers of osteolytic bone metastasis. Cancer Cell 24:542–556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Yang S, Zhang W, Cai M, Zhang Y, Jin F, Yan S, Baloch Z, Fang Z, Xue S, Tang R, Xiao J, Huang Q, Sun Y, Wang X (2018) Suppression of bone resorption by miR-141 in aged rhesus monkeys. J Bone Miner Res 33(10):1799–1812 [DOI] [PubMed] [Google Scholar]
  • 72.Min S, Liang X, Zhang M, Zhang Y, Mei S, Liu J, Liu J, Su X, Cao S, Zhong X, Li Y, Sun J, Liu Q, Jiang X, Che Y, Yang R (2013) Multiple tumor-associated microRNAs modulate the survival and longevity of dendritic cells by targeting YWHAZ and Bcl2 signaling pathways. J Immunol 190(5):2437–2446 [DOI] [PubMed] [Google Scholar]
  • 73.Chen C, Cheng P, Xie H, Zhou HD, Wu XP, Liao EY, Luo XH (2014) MiR-503 regulates osteoclastogenesis via targeting RANK. J Bone Miner Res 29(2):338–347 [DOI] [PubMed] [Google Scholar]
  • 74.Cao X (2011) Targeting osteoclast-osteoblast communication. Nat Med 17(11):1344–1346 [DOI] [PubMed] [Google Scholar]
  • 75.Negishi-Koga T, Shinohara M, Komatsu N, Bito H, Kodama T, Friedel RH, Takayanagi H (2011) Suppression of bone formation by osteoclastic expression of semaphorin 4D. Nat Med 17(11): 1473–1480 [DOI] [PubMed] [Google Scholar]
  • 76.Hayashi M, Nakashima T, Taniguchi M, Kodama T, Kumanogoh A, Takayanagi H (2012) Osteoprotection by semaphorin 3A. Nature. 485(7396):69–74 [DOI] [PubMed] [Google Scholar]
  • 77.Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, Suda T, Matsuo K (2006) Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab 4(2):111–121 [DOI] [PubMed] [Google Scholar]
  • 78.Takeshita S, Fumoto T, Matsuoka K, Park KA, Aburatani H, Kato S, Ito M, Ikeda K (2013) Osteoclast-secreted CTHRC1 in the coupling of bone resorption to formation. J Clin Invest 123(9):3914–3924 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Moverare-Skrtic S, Henning P, Liu X, Nagano K, Saito H, Borjesson AE, Sjogren K, Windahl SH, Farman H, Kindlund B et al. (2014) Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures. Nat Med 20(11): 1279–1288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D (2016) Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell 30(6):836–848 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Li D, Liu J, Guo B, Liang C, Dang L, Lu C, He X, Cheung HY, Xu L, Lu C et al. (2016) Osteoclast-derived exosomal miR-214–3p inhibits osteoblastic bone formation. Nat Commun 7(10872) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Liang C, Guo B, Wu H, Shao N, Li D, Liu J, Dang L, Wang C, Li H, Li S, Lau WK, Cao Y, Yang Z, Lu C, He X, Au DWT, Pan X, Zhang BT, Lu C, Zhang H, Yue K, Qian A, Shang P, Xu J, Xiao L, Bian Z, Tan W, Liang Z, He F, Zhang L, Lu A, Zhang G (2015) Aptamer-functionalized lipid nanoparticles targeting osteoblasts as a novel RNA interference-based bone anabolic strategy. Nat Med 21(3): 288–294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC, Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes TM, Hanauer A, Karsenty G (2004) ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry syndrome. Cell. 117(3):387–398 [DOI] [PubMed] [Google Scholar]
  • 84.Wang X, Guo B, Li Q, Peng J, Yang Z, Wang A, Li D, Hou Z, Lv K, Kan G, Cao H, Wu H, Song J, Pan X, Sun Q, Ling S, Li Y, Zhu M, Zhang P, Peng S, Xie X, Tang T, Hong A, Bian Z, Bai Y, Lu A, Li Y, He F, Zhang G, Li Y (2013) miR-214 targets ATF4 to inhibit bone formation. Nat Med 19(1):93–100 [DOI] [PubMed] [Google Scholar]
  • 85.Zhao C, Sun W, Zhang P, Ling S, Li Y, Zhao D, Peng J, Wang A, Li Q, Song J, Wang C, Xu X, Xu Z, Zhong G, Han B, Chang YZ, Li Y (2015) miR-214 promotes osteoclastogenesis by targeting Pten/PI3k/Akt pathway. RNA Biol 12(3):343–353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Jindra PT, Bagley J, Godwin JG, Iacomini J (2010) Costimulation-dependent expression of microRNA-214 increases the ability of T cells to proliferate by targeting Pten. J Immunol 185(2):990–997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Buckler JL, Walsh PT, Porrett PM, Choi Y, Turka LA (2006) Cutting edge: T cell requirement for CD28 costimulation is due to negative regulation of TCR signals by PTEN. J Immunol 177(7): 4262–4266 [DOI] [PubMed] [Google Scholar]
  • 88.Liu J, Li D, Dang L, Liang C, Guo B, Lu C, He X, Cheung HY, He B, Liu B et al. (2017) Osteoclastic miR-214 targets TRAF3 to contribute to osteolytic bone metastasis of breast cancer. Sci Rep 7(40487) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dambal S, Shah M, Mihelich B, Nonn L (2015) The microRNA-183 cluster: the family that plays together stays together. Nucleic Acids Res 43(15):7173–7188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ke K, Sul OJ, Rajasekaran M, Choi HS (2015) MicroRNA-183 increases osteoclastogenesis by repressing heme oxygenase-1. Bone. 81:237–246 [DOI] [PubMed] [Google Scholar]
  • 91.Stittrich AB, Haftmann C, Sgouroudis E, Kuhl AA, Hegazy AN, Panse I, Riedel R, Flossdorf M, Dong J, Fuhrmann F et al. (2010) The microRNA miR-182 is induced by IL-2 and promotes clonal expansion of activated helper T lymphocytes. Nat Immunol 11(11): 1057–1062 [DOI] [PubMed] [Google Scholar]
  • 92.Ichiyama K, Gonzalez-Martin A, Kim BS, Jin HY, Jin W, Xu W, Sabouri-Ghomi M, Xu S, Zheng P, Xiao C, Dong C (2016) The microRNA-183-96-182 cluster promotes T helper 17 cell pathogenicity by negatively regulating transcription factor Foxo1 expression. Immunity. 44(6):1284–1298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Inoue K, Deng Z, Chen Y, Giannopoulou E, Xu R, Gong S, Greenblatt MB, Mangala LS, Lopez-Berestein G, Kirsch DG, Sood AK, Zhao L, Zhao B (2018) Bone protection by inhibition of microRNA-182. Nat Commun 9(1):4108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP (2011) A ceRNA hypothesis: the Rosetta stone of a hidden RNA language? Cell. 146(3):353–358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A, Bozzoni I (2011) A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147(2):358–369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mohammed MA, Syeda JTM, Wasan KM, Wasan EK (2017) An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics. 9(4) [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES