Next Article in Journal
Evaluation of Commercially Available Viral Transport Medium (VTM) for SARS-CoV-2 Inactivation and Use in Point-of-Care (POC) Testing
Next Article in Special Issue
Small RNA Plays Important Roles in Virus–Host Interactions
Previous Article in Journal
Competency of Amphibians and Reptiles and Their Potential Role as Reservoir Hosts for Rift Valley Fever Virus
Previous Article in Special Issue
Disruption of Zika Virus xrRNA1-Dependent sfRNA1 Production Results in Tissue-Specific Attenuated Viral Replication
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of MicroRNAs in Bone Pathology during Chikungunya Virus Infection

by
Enakshi Roy
1,
Siddappa N. Byrareddy
2,3,4,* and
St Patrick Reid
1,*
1
Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
2
Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
3
Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
4
Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
*
Authors to whom correspondence should be addressed.
Viruses 2020, 12(11), 1207; https://doi.org/10.3390/v12111207
Submission received: 27 August 2020 / Revised: 10 October 2020 / Accepted: 19 October 2020 / Published: 23 October 2020
(This article belongs to the Special Issue MicroRNAs and Other Small RNAs in Viral Infections)

Abstract

:
Chikungunya virus (CHIKV) is an alphavirus, transmitted by mosquitoes, which causes Chikungunya fever with symptoms of fever, rash, headache, and joint pain. In about 30%–40% of cases, the infection leads to polyarthritis and polyarthralgia. Presently, there are no treatment strategies or vaccine for Chikungunya fever. Moreover, the mechanism of CHIKV induced bone pathology is not fully understood. The modulation of host machinery is known to be essential in establishing viral pathogenesis. MicroRNAs (miRNAs) are small non-coding RNAs that regulate major cellular functions by modulating gene expression. Fascinatingly, recent reports have indicated the role of miRNAs in regulating bone homeostasis and altered expression of miRNAs in bone-related pathological diseases. In this review, we summarize the altered expression of miRNAs during CHIKV pathogenesis and the possible role of miRNAs during bone homeostasis in the context of CHIKV infection. A holistic understanding of the different signaling pathways targeted by miRNAs during bone remodeling and during CHIKV-induced bone pathology may lead to identification of useful biomarkers or therapeutics.

1. Background

Chikungunya virus (CHIKV) is a positive-sense, single-stranded RNA virus of the Togaviridae family and Alphavirus genus [1]. The genome is 11.8 kb in length and has two open reading frames, the 5′ORF which encodes the non-structural proteins nsP1, nsP2, nsP3, and nsP4, and the 3′ORF, which encodes the structural proteins, capsid (C), envelope (E1 and E2), and two peptides (E3 and 6K). The virus was first isolated in 1952 from the Makonde plateau in southern Tanzania [2,3]. Since its initial outbreak in the mid-1900s, there have been numerous outbreaks of CHIKV infection in Africa. In the late 1950s and early 1960s, there were outbreaks of the disease in Thailand which affected about 31% of the population. In India, CHIKV was first detected in 1963 in West Bengal which was followed by several other outbreaks between 1964 and 1973. In 2004, there was a CHIKV outbreak in Kenya. In 2005, the largest CHIKV outbreak occurred in India which resulted in about 1.5 million infections. Another CHIKV outbreak occurred in 2005 in Réunion island which affected approximately one-third of the island’s population. Other cases of large-scale epidemics were in Mauritius in 2006, Gabon in 2007, and Thailand in 2008. In 2010, an autochthonous transmission of CHIKV was recorded in southern France. In 2011, an outbreak occurred in the Republic of Congo. Beginning in 2013, this virus spread to the Americas, in part due to travel from affected regions [4,5,6]. In 2014, outbreaks occurred in Martinique-Guadeloupe and French Polynesia. To date, three CHIKV genotypes have been identified, which are West African, Asian, and the East Central and South Asian (ECSA) lineages [5]. The sub Indian Ocean lineage (IOL) is a descendant of the ECSA lineage. CHIKV is mainly transmitted by Aedes (Ae.) species mosquitoes [4]. The word “chikungunya” means “that which bends up” in Makonde language to describe the stooped posture of infected patients due to musculoskeletal pain. CHIKV causes CHIK fever (CHIKF), a disease that is typically accompanied by fever, headache, rash, and arthralgia [7]. Although the mortality rate is low, the morbidity rate is high with 30%–40% of the infected patients developing a chronic stage characterized by debilitating arthritis similar in pathology with rheumatoid arthritis (RA) [7,8]. In rare cases, CHIKV infection also exhibit encephalitic pathology [9,10]. However, the bone related pathologies such as joint pain and arthritis-like conditions are major concerns of CHIKV infection.
MicroRNAs (miRNAs) are a class of small non-coding single stranded RNAs of 21–25 nucleotides in length which function as post-transcriptional regulators of gene expression [11,12]. miRNAs regulate gene expression by binding to the complementary 3′ untranslated regions (3′ UTR) of target mRNAs and inhibiting their translation either by mRNA degradation or translational repression [13]. Recently, miRNAs have been shown to bind to the 5′ UTR of target genes [14]. The miRNA mediated silencing of the mRNA targets occurs in cytoplasmic processing bodies (P-bodies) [15]. The formation of mature miRNAs is a sequential process. The miRNA gene is transcribed into primary miRNA (pri-miRNA) which is processed into precursor miRNA (pre-miRNA) by a DROSHA/DGCR8 complex [16,17,18]. The pre-miRNA is then exported to the cytoplasm where it undergoes further processing by DICER/TRBP/PACT enzyme complex to generate short double-stranded (ds) RNA [19]. Usually, the RNA strand with lower stability at the 5′ end is integrated into the RNA-induced silencing complex (RISC) and become a mature miRNA, while the strand with higher stability at the 5′ end is degraded [20]. The miRNA-induced silencing complex (miRISC) then binds to the 3′ UTR or 5′ UTR of the target mRNA, thus inhibiting its translation. [14]. By affecting gene expression, miRNAs regulate essential physiological and cellular processes including cell cycle, differentiation, proliferation, apoptosis, and immune response [13].
Viruses can alter host miRNA levels and the altered expression of those host miRNAs have been associated with the establishment of viral pathogenesis [21]. Here in this review, we highlight the recent studies that have indicated the association of host miRNA with CHIKV infection.

2. Role of miRNAs during CHIKV Infection

Viruses are known to hijack host gene expression and modulate fundamental cellular processes to establish infection and pathogenesis. Many studies have demonstrated the role of miRNAs during viral infections [21,22]. Besides having an imperative role in physiological functions, the aberrant expression of miRNAs is associated with pathogenesis of various diseases including viral infections [21,22,23]. Recently, altered expression of several miRNAs (listed in Table 1) has been observed during CHIKV infection [24,25,26,27,28,29,30,31,32].
Given its significance in the regulation of cellular processes, we focus on the recent reports that elucidate the expression levels of different host miRNAs, related target genes, their functions in cellular processes, and role in CHIKV pathogenesis.

2.1. Antiviral Role of miRNAs

The generation of antiviral miRNAs has been observed in many viral infections [21,22]. Computational analysis showed a number of significantly modulated miRNAs in early CHIKV infection are involved in apoptosis and JAK-STAT signaling pathways [32]. Interestingly, the JAK/STAT pathway is known to be one of the key signaling pathways in the interferon (IFN) response against viral infection [33]. Reverse genetic approaches and functional studies in Ae. aegypti mosquitoes revealed that increased resistance to Dengue virus (DENV) and Zika virus (ZIKV) infections is mediated by the JAK/STAT pathway [34]. Moreover, CHIKV non-structural protein 2 (nsP2), has been associated with the JAK/STAT pathway [35]. Thus, evaluating the interaction among viral proteins, miRNAs and their involvement in the JAK-STAT pathway holds potential for exploratory studies in CHIKV pathogenesis.
miRNA profiling in CHIKV-infected human skin fibroblasts showed differential expression of a number of miRNAs in the early stage of CHIKV infection [26]. The miRNAs were predicted to target immune-related signaling pathways including JAK/STAT, MAPK, WNT, and retinoic acid inducible gene I (RIG-I)-like receptor pathways [26]. Interestingly, both JAK/STAT and MAPK pathways have been associated with CHIKV infection [35,36]. Additionally, the WNT signaling pathway can regulate IFN response in flaviviruses [37]. The expressions of hsa-miR-15 and hsa-miR-16 were altered during CHIKV infection [26]. In normal physiology, a number of cellular processes are regulated by hsa-miR-15 and hsa-miR-16 and altered expression of these miRNAs is observed in many other viral infections and diseases [38,39,40,41,42]. Interestingly, hsa-miR-15 and hsa-miR-16 play important roles in inducing apoptosis by targeting the anti-apoptotic protein BCL2 [43]. Additionally, downregulated expression of hsa-miR-15 was found in arthritic synovial tissue, whereas hsa-miR-16 level was high in sera of RA patient [38,44]. rno-miR-32–5 p is a negative regulator of phosphatase and tensin homolog (PTEN) [45]. Thus, understanding the functional relevance of these miRNAs during CHIKV infection would be helpful for the development of novel drug targets.
Microarray analysis in CHIKV infected HEK293T cells revealed a set of 152 differentially regulated miRNAs [28]. Among these, about 65%–70% of the differentially regulated miRNAs were significantly upregulated and the remaining were downregulated. RT-PCR analysis showed that among the upregulated hsa-miRNAs, hsa-miR-744, hsa-miR-638, and hsa-miR-503 were significantly upregulated. Interestingly, 53% of the observed upregulated miRNAs and 45% of the downregulated miRNA were altered in other viral infections including hepatitis B virus (HBV), hepatitis C virus (HCV), human papillomavirus (HPV), and human immunodeficiency virus (HIV). Further analysis of the miRNA pattern demonstrated that the altered miRNAs were members of different miRNA cluster including hsa-miR-17-92, let-7e/99b, hsa-miR-191/425, hsa-miR-106b/25, hsa-miR-23a/24, and hsa-miR-15b/16 clusters which further indicated that these miRNA clusters are co-regulated in response to CHIKV infection [28]. The pathway analysis predicted TGF-β, WNT pathway, endocytosis ubiquitin mediated proteolysis, proteasome and lysosome associated genes, and the cell cycle pathways as targets of the altered miRNAs [28]. Moreover, qRT-PCR results confirmed that the altered miRNAs induced TGF-β genes (mothers against decapentaplegic homolog 6 (SMAD6), JUN, and ski-like protein (SKIL) genes) and endocytosis pathway genes (C-X-C motif chemokine receptor 4 (CXCR4), heat shock cognate 71 kDa protein (HSPA8), adrenoceptor beta 1 (ADRB1)), but inhibited genes involved in cell cycle pathways (cell division cycle 27 (CDC27) and CDC23). The cells treated with a TGF-β inhibitor, SB-431542, showed increased CHIKV mediated cell death compared to untreated cells, indicating that TGF-β production is involved in regulating CHIKV infection [28]. TGF-β signaling pathway is a key network in regulating important cellular processes including proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and migration [46]. Studies show that this pathway is modulated during many viral infections [47]. Importantly, TGF-β pathway is a significant factor in age related complications during CHIKV infection [48]. Thus, miRNA mediated regulation of TGF-β pathway may contribute to host response against CHIKV infection.

2.2. Pro-Viral Role of miRNAs

The ability of miRNAs to regulate gene expression makes them particularly useful for viruses. Often viruses employ cellular miRNAs to target specific genes and downregulate their expression to establish infection [21,22]. The expression of hsa-miR-146a was found to be upregulated in CHIKV-infected human synovial fibroblasts where TRAF6 and IRAK1 were predicted as targets (Table 1) [24]. The expression of these targets was restored in cells transfected with hsa-anti-miR-146a. In addition, overexpression of hsa-miR-146a leads to decreased phosphorylation of NF-кB during infection [24]. The conclusions were similar to another finding which demonstrated that increased expression of hsa-miR-146a enhanced DENV replication by targeting the TRAF6-mediated NF-кB pathway [49]. These results suggested a role of hsa-miR-146a-mediated targeting of the NF-кB pathway during CHIKV pathogenesis.
In human synovial fibroblasts, an miRNA microarray identified a subset of 26 differentially expressed miRNAs (DEMs) during CHIKV infection (Table 1) [25]. Among the DEMS, expression of hsa-miR-4717-3p, hsa-miR-4299, hsa-miR-1264, and hsa-miR-21-5p were significantly upregulated. AKT3 was predicted as a target for hsa-miR-4717-3p (Table 1). The AKT3 protein is a key regulator of the PI3K/AKT/mTOR signaling pathway which influences various cellular processes including metabolism, growth, proliferation, survival, transcription, and protein synthesis [50]. Moreover, dysregulation of the PI3K/AKT/mTOR pathway has been implicated in many diseases [51,52]. Interestingly, this pathway is moderately activated during CHIKV infection [53]. Thus, hsa-miR-4717-3p may mediate suppression of the robust inflammatory response during CHIKV infection, by targeting AKT3 [25]. In response to an infection, the host cellular system elicits a cytokine mediated immune response, but often due to inefficient pathogen clearance, the immune response results in inflammation [54]. The expression of hsa-miR-4299 was upregulated during CHIKV infection and the suppressor of cytokine signaling 7 protein (SOCS7) was predicted as the target (Table 1) [25]. SOCS7 is known to negatively regulate the STAT3 protein which can either induce IL-6-mediated inflammation or IL-10-mediated suppression of inflammation during CHIKV infection [55]. STAT3 can also promote viral replication and persistence [56]. Thus, hsa-miR-4299 could mediate suppression of SOCS7 which may result in increased STAT3 expression contributing to suppressed immune response during CHIKV infection. As earlier mentioned, viruses employ different strategies to exploit cellular pathways for optimizing chances of survival [57,58]. Agrawal et al. showed that expression of hsa-miR-1264 increased during CHIKV infection and TRIM26 was predicted to be its target (Table 1) [25]. TRIM proteins function as E3 ubiquitin ligase playing an important role in antiviral responses through ubiquitination and proteasomal degradation of IRF3 genes during viral infections [59]. Thus, this indicates that CHIKV infection may lead to hsa-miR-1264-mediated suppression of TRIM26 resulting in depleted antiviral response and enhanced viral replication and persistence [25]. Another E3 ubiquitin ligase, PELI1 was targeted by hsa-miR-21-5p whose expression was upregulated during infection (Table 1) [25]. PELI1 can suppress the NF-кB pathway by ubiquitination and degradation of an NF-кB-inducing kinase (NIK) [60]. As the NF-кB pathway plays a critical role in antiviral response, thus, increased expression of miR-21 during CHIKV infection may contribute to suppression of cytokine signaling by modulating the NF-кB pathway.
A genome-wide miRNA screen using high throughput RNA sequencing in Huh-7.5.1 cells revealed that alphaviruses have a binding site for hsa-miR-124 [27]. A significant increase in CHIKV production was observed on overexpressing hsa-miR-124, whereas inhibiting hsa-miR-124 led to reduced CHIKV infection. In rare cases, CHIKV infection can result in encephalitic symptoms [9,10]. hsa-miR-124 is predominantly found in neurons and act as a key negative regulator of neuroinflammation [61]. An altered expression of hsa-miR-124 has been associated with brain disease [61]. Thus, it would be interesting to evaluate whether hsa-miR-124 is associated with encephalitic pathology during CHIKV infection. In a study by Nakamachi et al., decreased expression of hsa-miR-124 was observed in fibroblast like synoviocytes (FLS) of patients with RA where hsa-miR-124a contributed to the inflammatory processes in RA pathogenesis by targeting the monocyte chemoattractant protein-1 (MCP-1) and cyclin-dependent kinase-2 (CDK-2) [62]. Thus, hsa-miR-124 may have a role in contributing to inflammation observed during CHIKV infection.

2.3. Aberrant Expression of miRNAs in Mosquito Cells during CHIKV Infection

To establish infection and increase virus survival in a mosquito vector, viruses modify the transcriptional profile of the vector [63]. In Aag-2 cells, aae-miR-2944b-5p and aae-miR-2b were observed to have binding sites for the 3′ UTR of CHIKV [29]. When mosquitoes were treated with antagomiR-2944b-5p, they showed more susceptibility to CHIKV infection compared to untreated control which suggested the role of antagomiR-2944b-5p in viral replication. The host vacuolar protein sorting-associated protein 13 (VPS-13) was predicted as a target of aae-miR-2944b-5p. In Ae. aegypti, VPS-13 functions in maintaining the mitochondrial membrane potential (MtMP) [29]. Interestingly, studies report that host mitochondria are involved in combating the oxidative stress induced during viral infections [64]. Silencing aae-miR-2944b-5p in Aag-2 cells and infecting with CHIKV increased cellular MtMP, which indicated that aae-miR-2944b-5p interacts with VPS-13 to maintain MtMP [29]. In humans, VPS-13 is involved in post Golgi apparatus sorting and trafficking. Thus, studying the effect of hsa-miR-2944b-5p on VPS-13 expression in human cell lines during CHIKV infection can lead to identification of novel drug target.
Using next generation RNA sequencing, the expressions of a set of eight miRNAs were found to be altered during CHIKV infection [30]. Among them, the expressions of aae-miR-100, aae-miR-283, aae-miR-305-3p, and aae-miR-927 were significantly upregulated and the expressions of aae-miR-1000, aae-miR-2b, aae-miR-2c-3p, and aae-miR-190-5p were downregulated. Target prediction revealed that aae-miR-100, aae-miR-283, and aae-miR-305-3p commonly affected NK cell-mediated cytotoxicity and protein processing in ER pathways. The analysis also revealed that the metabolic pathways such as the TCA cycle, dorso-ventral axis formation, and valine, leucine, and isoleucine degradation pathways were affected by aae-miR-100 and aae-miR-305-3p. aae-amiR-927 and aae-miR-305-3p were predicted to target SNARE interactions in vesicular transport. Among these, aae-miR-305-3p was predicted to target pathways essential for viral entry such as ECM receptor interaction, endocytosis, and SNARE interactions in vesicular transport. The downregulated aae-amiR-1000, aae-miR-2b, and aae-miR-2c targeted the ribosomal pathway. The upregulated miRNAs targeted genes which encodes for protein tyrosine phosphatase SHP2, ERK1/2, and ubiquitin fusion degradation protein, respectively, whereas the downregulated miRNAs targeted the gene that encodes for the 40S ribosomal protein S16. In another study, next-generation sequencing identified the altered expression of 13 miRNAs during CHIKV infection in Aag-2 cells [31]. Target prediction analysis showed aae-miR-2b targets URM and ubiquitin whereas aae-miR-100 targets CDC42 and sumo-ligase. When cells were treated with aae-antagomiR-2b, increased CHIKV replication was observed. The expression of URM was also significantly high in CHIKV infected cells. Furthermore, CHIKV replication was reduced to 50% in URM knock down cells, indicating that aae-miR-2b-mediated regulation of URM plays a significant role in chikungunya replication.
Usually in mosquito vectors, viruses establish infection in the salivary gland during a blood meal [63]. For establishing a successful infection, viruses often modulate the gene expression of several proteins in the salivary gland [63]. Next generation sequencing showed that aae-miR-bantam, aae-miR-263a, aae-miR-125, and aae-miR-285 were significantly upregulated in CHIKV-infected Ae. aegypti saliva [65]. In Ae. albopictus saliva, aal-miR-43b, aal-miR-43a, aal-miR-413a, aal-miR-5, and aal-miR-249 were upregulated [65]. In addition, Aag-2 cells and BHK-21 cells showed decreased CHIKV titers when treated with inhibitors against selected miRNAs indicating the role of salivary gland miRNAs in modulating CHIKV replication. Another study predicted a set of miRNAs that commonly targeted the different genotypes of CHIKV where aae-miR-282-5p, aae-miR-34-3p, and aae-miR-11-5p had binding sites for CHIKV [66]. Moreover, aae-miR-11-5p was conserved among the different lineages of CHIKV and was predicted to target the end of subgenomic untranslated RNA region, thus, indicating that the CHIKV structural proteins may regulate the formation of a miRNA-viral RNA (vRNA) complex at the end of subgenomic RNA untranslated regions, thereby preventing the binding of host translational factors on vRNA.

3. Possible Role of miRNAs in Bone Homeostasis in the Context of CHIKV Infection

CHIKV infection is associated with bone pathology and it was first indicated by the presence of bony lesions in CHIKV-infected IRF 3/7-/- mice [67]. In CHIKV-infected patients, MRI results showed the presence of erosive arthritis [68]. Bone is one of the most dynamic organs in the body that continuously undergoes remodeling. Bone homeostasis is a highly regulated and complex process involving a fine balance between osteoblastogenesis and osteoclastogenesis [69]. Osteoblastogenesis is the process of bone formation which results from differentiation of mesenchymal stem cells (MSCs) into osteoblastic cell lineage forming the bone cells or osteoblasts (OBs) and later into osteocytes, the mature OBs [70]. Conversely, osteoclastogenesis is the process of bone resorption where the formation of multinucleated osteoclasts (OCs) occurs from the fusion of myeloid precursors which arise by differentiation of hematopoietic stem cells (HSCs) [71]. Many complex processes, signaling pathways, and transcription factors govern osteoblastogenesis and osteoclastogenesis in maintaining normal bone homeostasis (Figure 1 and Figure 2).
During osteoblastogenesis, the key signaling pathways activated are canonical WNT, NOTCH, Hedgehog, BMP, SMAD, MAPK, and the receptor activator of nuclear factor kβ (RANK), osteoprotegerin (OPG)-and RANK ligand (RANKL) [72,73]. These pathways result in expression of the key transcription factors identified during osteoblastogenesis which are the runt-related transcription factor 2 (RUNX2), and Osterix (OSX) [74,75]. However, there are other transcription factors that also function in bone homeostasis [76,77]. These transcription factors subsequently induce the expression of other osteogenic genes including alkaline phosphatase (ALP), type I collagen (COl-I), osteocalcin (OCN), osteonectin (ON), and bone sialoprotein (BSP) [76,77,78]. Similarly, processes such as OC differentiation from myeloid precursors, maturation, and survival of the OC are regulated by a variety of environmental factors including cytokines, growth factors, and hormones which influence the RANK-RANKL, MAPK, PI3K/AKT, and NF-kβ pathways [72,73]. These signaling pathways in turn regulate the expression of various transcription factors among which nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1) is critical [79,80]. NFATC1 is the major regulator of the early phase of osteoclastogenesis, which induces the expression of other osteoclastic genes in the late phase such as tartrate-resistant acid phosphatase (TRAP), cathepsin k (CTSK), and dendrocyte expressed seven transmembrane proteins (DCSTAMP) [79,81,82,83,84,85]. Conversely, few transcription factors can also negatively regulate osteoclastogenesis [86,87,88,89,90,91].
Bone homeostasis is regulated by an array of transcription factors and complex signaling pathways; thus, understanding how these pathways are modulated during CHIKV infection is of great importance. Recently, a number of studies indicated that miRNAs can regulate both osteoclastogenesis and osteoblastogenesis thereby acting as fine modulators of bone homeostasis [92] (Table 2 and Table 3) (Figure 1 and Figure 2).
The levels of miRNAs are altered in many bone-related pathological conditions [142]. Furthermore, as mentioned earlier, dysregulated expression of miRNAs has been reported during CHIKV infection (Table 1). Thus, understanding the specific roles of miRNAs in bone homeostasis during CHIKV infection will be critical for the identification of novel biomarkers in altered bone homeostasis during infection and for the development of miRNA-based therapeutics.
In osteogenically differentiated MSCs derived from dental and craniofacial tissues, the expression of hsa-miR-21 was down-regulated, and its overexpression suppressed osteoblastogenesis [94]. SMAD5, the upstream regulator of RUNX2 during osteogenesis, was the target of hsa-miR-21 [94]. However, in mouse osteoblast MC3T3-E1 cells, mmu-miR-21 induced osteogenic differentiation by targeting SMAD7 [95]. Sun et al., also showed that mmu-miR-21 induced osteogenesis as overexpression of mmu-miR-21 resulted in increased mineralization and bone healing properties in a femur fracture model in rats [143]. During CHIKV infection, an upregulated expression of hsa-miR-21-5p was observed, and PELI1, a E3 ubiquitin protein ligase, was predicted as the target [25]. PELI1 has been shown to inhibit the NF-κB signaling pathway, which is an important pathway during osteoclastogenesis [60]. Thus, further studies may be conducted to investigate whether miR-21 can impair osteoclastogenesis during CHIKV infection. FAK signaling pathway acts as a critical signaling pathway in the early stages of osteoblastogenesis [144]. During CHIKV infection, hsa-miR-138-2-3p is upregulated and a number of genes are predicted as targets including MAPK13 [145]. MAPK13 encodes p38 MAPK which plays an important role in bone homeostasis [146]. hsa-miR-138 is also downregulated during osteoblastogenesis as it can target FAK and inhibit the FAK-mediated signaling pathway [145]. Additionally, suppression of hsa-miR-138 expression with antagomiR-138 increased ectopic bone formation in vivo and overexpression of hsa-miR-138 reversed the effects, thus indicating that hsa-miR-138 impairs osteogenic differentiation by targeting FAK and its downstream signaling pathways. However, whether miR-138 regulates any FAK-mediated MAPK-signaling pathway during CHIKV infection is not yet known. Many reports have suggested that joint inflammation is associated with arthritic-like symptoms during CHIKV infection [147]. hsa-miR-146 has been associated with many viral and microbial infections and also with inflammatory conditions such as RA [49,148,149]. An upregulated expression of hsa-miR-146a was observed in synovial fibroblasts during CHIKV infection [24]. Furthermore, TRAF6 and IRAK1 were predicted as targets of hsa-miR-146a [25]. It is known that during viral infections, TRAF6 and IRAK1 activate the NF-кB signaling pathway to produce pro-inflammatory cytokines for combating infection [49]. Additionally, the NF-кB signaling pathway has been shown to play an important role during osteoclastogenesis. However, the effect of hsa-miR-146a on the NF-кB signaling pathway during CHIKV infection remains unknown.

4. Conclusions

Understanding the involvement of miRNAs during bone homeostasis in the context of CHIKV infection is of much interest as identification of novel biomarkers and/or development of miRNA-based therapeutics against viral infections is a promising area of research. Several miRNAs already serve as biomarkers and have been associated with pathologies, stages, and/or progression of different diseases. However, the use of miRNAs as biomarkers to diagnose viral diseases is still uncommon. Recently, hsa-miR-181c-5p and hsa-miR-1254 were identified as biomarkers for detection of H1N1 virus influenza [150]. In miRNA-based therapeutics, the developed miRNA either targets the pathogen or host factor during infection. During CHIKV pathogenesis, the two broad areas that can be targeted for drug development are (1) to directly impact virus replication or (2) to modulate host factors to mitigate arthritic-like symptoms caused due to infection. At present, a number of bioinformatic databases and high throughput screens are available to predict miRNA targets during preclinical therapeutic investigations. Additionally, a variety of in vitro cell culture models and in vivo mouse and non-human primate models are available to investigate the efficacy, toxicity, and safety of miRNA therapeutics. A phase 2 clinical trial with miravirsen (locked nucleic acid–modified DNA phosphorothioate antisense oligonucleotide that sequesters the mature hsa-miR-122 in a stable heteroduplex, thereby suppressing its function) in chronic hepatitis C virus (HCV)-infected patients showed reduced HCV RNA levels that persisted beyond the end of active therapy [151]. Another product, RG-101 (an N-acetyl-D-galactosamine-conjugated RNA antagomiR that targets hsa-miR-122 in HCV infected hepatocytes), was used in a clinical trial, which also resulted in undetectable HCV RNA in patients; however, it produced adverse effects due to which the trial was put on hold [152]. Thus, the transition of laboratory findings to clinical applications of miRNA-based diagnostics and therapeutics still remains a challenge and warrants further research.

Funding

This work was supported by NIH/NIAID 1R21AI140026-01.

Acknowledgments

All the figures were created using Biorender.com. We would like to thank Opeoluwa O. Oyewole and M. Jane Morwitzer for their valuable comments.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Morrison, C.R.; Plante, K.S.; Heise, M.T. Chikungunya Virus: Current Perspectives on a Reemerging Virus. Microbiol. Spectr. 2016, 4, 143–161. [Google Scholar]
  2. Robinson, M.C. An epidemic of virsus disease in Southern Province, Tanganyika Territory, in 1952–53, Clinical Features. Trans. R. Soc. Trop. Med. Hyg. 1955, 49, 28–32. [Google Scholar] [CrossRef]
  3. Lumsden, W.H.R. An epidemic of virus disease in Southern Province, Tanganyika Territory, in 1952–53. II. General description and epidemiology. Trans. R. Soc. Trop. Med. Hyg. 1955, 49, 33–57. [Google Scholar] [CrossRef]
  4. Zeller, H.; Van Bortel, W.; Sudre, B. Chikungunya: Its History in Africa and Asia and Its Spread to New Regions in 2013–2014. J. Infect. Dis. 2016, 214, S436–S440. [Google Scholar] [CrossRef]
  5. Weaver, S.C.; Forrester, N.L. Chikungunya: Evolutionary history and recent epidemic spread. Antivir. Res. 2015, 120, 32–39. [Google Scholar] [CrossRef]
  6. Petersen, L.R.; Powers, A.M. Chikungunya: Epidemiology. F1000 Res. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
  7. Cunha, R.V.D.; Trinta, K.S. Chikungunya virus: Clinical aspects and treatment–A Review. Mem. Inst. Oswaldo Cruz. 2017, 112, 523–531. [Google Scholar] [CrossRef]
  8. Ganesan, V.K.; Duan, B.; Reid, S.P. Chikungunya Virus: Pathophysiology, Mechanism, and Modeling. Viruses 2017, 9, 368. [Google Scholar] [CrossRef] [Green Version]
  9. Rocha, V.F.D.; de Oliveira, A.H.P.; Bandeira, A.C.; Sardi, S.I.; Garcia, R.F.; Magalhães, S.A.; Sampaio, C.A.; Campos Soares, G. Chikungunya Virus Infection Associated with Encephalitis and Anterior Uveitis. Ocul. Immunol. Inflamm. 2018, 26, 677–679. [Google Scholar] [CrossRef]
  10. Mehta, R.; Gerardin, P.; Brito CAAd Soares, C.N.; Ferreira, M.L.B.; Solomon, T. The neurological complications of chikungunya virus: A systematic review. Rev. Med. Virol. 2018, 28, e1978. [Google Scholar] [CrossRef] [Green Version]
  11. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
  12. Ambros, V. The functions of animal microRNAs. Nature 2004, 431, 350–355. [Google Scholar] [CrossRef] [PubMed]
  13. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  14. Lee, I.; Ajay, S.S.; Yook, J.I.; Kim, H.S.; Hong, S.H.; Kim, N.H.; Dhanasekaran, S.M.; Chinnaiyan, A.M.; Athey, B.D. New class of microRNA targets containing simultaneous 5’-UTR and 3’-UTR interaction sites. Genome Res. 2009, 19, 1175–1183. [Google Scholar] [CrossRef] [Green Version]
  15. Liu, J.; Valencia-Sanchez, M.A.; Hannon, G.J.; Parker, R. MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nat. Cell Biol. 2005, 7, 719–723. [Google Scholar] [CrossRef] [Green Version]
  16. Han, J.; Lee, Y.; Yeom, K.H.; Kim, Y.K.; Jin, H.; Kim, V.N. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004, 18, 3016–3027. [Google Scholar] [CrossRef] [Green Version]
  17. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  18. Murchison, E.P.; Hannon, G.J. miRNAs on the move: miRNA biogenesis and the RNAi machinery. Curr. Opin. Cell Biol. 2004, 16, 223–229. [Google Scholar] [CrossRef]
  19. Lund, E.; Dahlberg, J.E. Substrate Selectivity of Exportin 5 and Dicer in the Biogenesis of MicroRNAs. Cold Spring Harb. Symp. Quant. Biol. 2006, 71, 59–66. [Google Scholar] [CrossRef] [Green Version]
  20. Rana, T.M. Illuminating the silence: Understanding the structure and function of small RNAs. Nat. Rev. Mol. Cell Biol. 2007, 8, 23–36. [Google Scholar] [CrossRef]
  21. Girardi, E.; Lopez, P.; Pfeffer, S. On the Importance of Host MicroRNAs During Viral Infection. Front. Genet. 2018, 9, 439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bruscella, P.; Bottini, S.; Baudesson, C.; Pawlotsky, J.M.; Feray, C.; Trabucchi, M. Viruses and miRNAs: More Friends than Foes. Front. Microbiol. 2017, 8, 824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ardekani, A.M.; Naeini, M.M. The Role of MicroRNAs in Human Diseases. Avicenna J. Med. Biotechnol. 2010, 2, 161–179. [Google Scholar] [PubMed]
  24. Selvamani, S.P.; Mishra, R.; Singh, S.K. Chikungunya virus exploits miR-146a to regulate NF-kappaB pathway in human synovial fibroblasts. PLoS ONE 2014, 9, e103624. [Google Scholar] [CrossRef] [PubMed]
  25. Agrawal, M.; Pandey, N.; Rastogi, M.; Dogra, S.; Singh, S.K. Chikungunya virus modulates the miRNA expression patterns in human synovial fibroblasts. J. Med. Virol. 2020, 92, 139–148. [Google Scholar] [CrossRef] [PubMed]
  26. Parashar, D.; Paingankar, M.S.; More, A.; Patil, P.; Amdekar, S. Altered microRNA expression signature in Chikungunya-infected mammalian fibroblast cells. Virus Genes. 2018, 54, 502–513. [Google Scholar] [CrossRef] [PubMed]
  27. López, P.; Girardi, E.; Mounce, B.C.; Weiss, A.; Chane-Woon-Ming, B.; Messmer, M.; Kaukinen, P.; Kopp, A.; Bortolamiol-Becet, D.; Fendri, A.; et al. High-Throughput Fluorescence-Based Screen Identifies the Neuronal MicroRNA miR-124 as a Positive Regulator of Alphavirus Infection. J. Virol. 2020, 94, 2119–2145. [Google Scholar] [CrossRef]
  28. Saxena, T.; Tandon, B.; Sharma, S.; Chameettachal, S.; Ray, P.; Ray, A.R.; Kulshreshtha, R. Combined miRNA and mRNA signature identifies key molecular players and pathways involved in chikungunya virus infection in human cells. PLoS ONE 2013, 8, e79886. [Google Scholar] [CrossRef] [Green Version]
  29. Dubey, S.K.; Shrinet, J.; Sunil, S. Aedes aegypti microRNA, miR-2944b-5p interacts with 3’UTR of chikungunya virus and cellular target vps-13 to regulate viral replication. PLoS Negl. Trop Dis. 2019, 13, e0007429. [Google Scholar] [CrossRef]
  30. Shrinet, J.; Jain, S.; Jain, J.; Bhatnagar, R.K.; Sunil, S. Next generation sequencing reveals regulation of distinct Aedes microRNAs during chikungunya virus development. PLoS Negl. Trop. Dis. 2014, 8, e2616. [Google Scholar] [CrossRef]
  31. Dubey, S.K.; Shrinet, J.; Jain, J.; Ali, S.; Sunil, S. Aedes aegypti microRNA miR-2b regulates ubiquitin-related modifier to control chikungunya virus replication. Sci. Rep. 2017, 7, 17666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sharma, A.; Balakathiresan, N.S.; Maheshwari, R.K. Chikungunya Virus Infection Alters Expression of MicroRNAs Involved in Cellular Proliferation, Immune Response and Apoptosis. Intervirology 2015, 58, 332–341. [Google Scholar] [CrossRef]
  33. Nan, Y.; Wu, C.; Zhang, Y.J. Interplay between Janus Kinase/Signal Transducer and Activator of Transcription Signaling Activated by Type I Interferons and Viral Antagonism. Front. Immunol. 2017, 8, 1758. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, W.S.; Webster, J.A.; Madzokere, E.T.; Stephenson, E.B.; Herrero, L.J. Mosquito antiviral defense mechanisms: A delicate balance between innate immunity and persistent viral infection. Parasit Vectors 2019, 12, 165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Fros, J.J.; Liu, W.J.; Prow, N.A.; Geertsema, C.; Ligtenberg, M.; Vanlandingham, D.L.; Schnettler, E.; Vlak, J.M.; Suhrbier, A.; Khromykh, A.A.; et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J. Virol. 2010, 84, 10877–10887. [Google Scholar] [CrossRef] [Green Version]
  36. Varghese, F.S.; Thaa, B.; Amrun, S.N.; Simarmata, D.; Rausalu, K.; Nyman, T.A.; Merits, A.; McInerney, G.M.; Ng, L.F.P.; Ahola, T. The Antiviral Alkaloid Berberine Reduces Chikungunya Virus-Induced Mitogen-Activated Protein Kinase Signaling. J. Virol. 2016, 90, 9743–9757. [Google Scholar] [CrossRef] [Green Version]
  37. Smith, J.L.; Jeng, S.; McWeeney, S.K.; Hirsch, A.J. A MicroRNA Screen Identifies the Wnt Signaling Pathway as a Regulator of the Interferon Response during Flavivirus Infection. J. Virol. 2017, 91, e02388-16. [Google Scholar] [CrossRef] [Green Version]
  38. Li, G.; Qiu, Z. Deletion of miR-15 Protects Against Rheumatoid Arthritis via Deregulating its Target Gene BCL2L2 and Repressing NF-κB Pathway. Ann. Clin. Lab. Sci. 2019, 49, 581–589. [Google Scholar]
  39. Bhomia, M.; Sharma, A.; Gayen, M.; Gupta, P.; Maheshwari, R.K. Artificial microRNAs can effectively inhibit replication of Venezuelan equine encephalitis virus. Antivir. Res. 2013, 100, 429–434. [Google Scholar] [CrossRef]
  40. Gao, Z.; Dou, Y.; Chen, Y.; Zheng, Y. MicroRNA roles in the NF- kappaB signaling pathway during viral infections. Biomed. Res. Int. 2014, 2014, 436097. [Google Scholar] [CrossRef] [Green Version]
  41. Makkoch, J.; Poomipak, W.; Saengchoowong, S.; Khongnomnan, K.; Praianantathavorn, K.; Jinato, T.; Poovorawan, Y.; Payungporn, S. Human microRNAs profiling in response to influenza A viruses (subtypes pH1N1, H3N2, and H5N1). Exp. Biol. Med. 2016, 241, 409–420. [Google Scholar] [CrossRef] [Green Version]
  42. Hullinger, T.G.; Montgomery, R.L.; Seto, A.G.; Dickinson, B.A.; Semus, H.M.; Lynch, J.M.; Dalby, C.M.; Robinson, K.; Stack, C.; Latimer, P.A.; et al. Inhibition of miR-15 protects against cardiac ischemic injury. Circ. Res. 2012, 110, 71–81. [Google Scholar] [CrossRef]
  43. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Moran-Moguel, M.C.; Petarra-Del Rio, S.; Mayorquin-Galvan, E.E.; Zavala-Cerna, M.G. Rheumatoid Arthritis and miRNAs: A Critical Review through a Functional View. J. Immunol. Res. 2018, 2018, 2474529. [Google Scholar] [CrossRef] [PubMed]
  45. Zhu, G.; Chai, J.; Ma, L.; Duan, H.; Zhang, H. Downregulated microRNA-32 expression induced by high glucose inhibits cell cycle progression via PTEN upregulation and Akt inactivation in bone marrow-derived mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2013, 433, 526–531. [Google Scholar] [CrossRef]
  46. Fabregat, I.; Caballero-Diaz, D. Transforming Growth Factor-beta-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front. Oncol. 2018, 8, 357. [Google Scholar] [CrossRef] [Green Version]
  47. Mirzaei, H.; Faghihloo, E. Viruses as key modulators of the TGF-beta pathway; a double-edged sword involved in cancer. Rev. Med. Virol. 2018, 28, e1967. [Google Scholar] [CrossRef]
  48. Uhrlaub, J.L.; Pulko, V.; DeFilippis, V.R.; Broeckel, R.; Streblow, D.N.; Coleman, G.D.; Park, B.S.; Lindo, J.F.; Vickers, I.; Anzinger, J.J.; et al. Dysregulated TGF-beta Production Underlies the Age-Related Vulnerability to Chikungunya Virus. PLoS Pathog. 2016, 12, e1005891. [Google Scholar] [CrossRef] [Green Version]
  49. Nahand, J.S.; Karimzadeh, M.R.; Nezamnia, M.; Fatemipour, M.; Khatami, A.; Jamshidi, S.; Moghoofei, M.; Taghizadieh, M.; Hajighadimi, S.; Shafiee, A.; et al. The role of miR-146a in viral infection. IUBMB Life 2020, 72, 343–360. [Google Scholar] [CrossRef]
  50. Bartok, B.; Boyle, D.L.; Liu, Y.; Ren, P.; Ball, S.T.; Bugbee, W.D.; Rommel, C.; Firestein, G.S. PI3 kinase delta is a key regulator of synoviocyte function in rheumatoid arthritis. Am. J. Pathol. 2012, 180, 1906–1916. [Google Scholar] [CrossRef]
  51. Malemud, C.J. The PI3K/Akt/PTEN/mTOR pathway: A fruitful target for inducing cell death in rheumatoid arthritis? Future Med. Chem. 2015, 7, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  52. Smith, M.V.; Lee, M.J.; Islam, A.S.; Rohrer, J.L.; Goldberg, V.M.; Beidelschies, M.A.; Greenfield, E.M. Inhibition of the PI3K-Akt signaling pathway reduces tumor necrosis factor-alpha production in response to titanium particles in vitro. J. Bone Joint Surg. Am. 2007, 89, 1019–1027. [Google Scholar] [PubMed]
  53. Thaa, B.; Biasiotto, R.; Eng, K.; Neuvonen, M.; Götte, B.; Rheinemann, L.; Mutso, M.; Utt, A.; Varghese, F.; Balistreri, G.; et al. Differential Phosphatidylinositol-3-Kinase-Akt-mTOR Activation by Semliki Forest and Chikungunya Viruses Is Dependent on nsP3 and Connected to Replication Complex Internalization. J. Virol. 2015, 89, 11420–11437. [Google Scholar] [CrossRef] [Green Version]
  54. Rouse, B.T.; Sehrawat, S. Immunity and immunopathology to viruses: What decides the outcome? Nat. Rev. Immunol. 2010, 10, 514–526. [Google Scholar] [CrossRef]
  55. Yang, R.; Xu, X.; Li, H.; Chen, J.; Xiang, X.; Dong, Z.; Zhang, D. p53 induces miR199a-3p to suppress SOCS7 for STAT3 activation and renal fibrosis in UUO. Sci. Rep. 2017, 7, 43409. [Google Scholar] [CrossRef] [Green Version]
  56. Kuchipudi, S.V. The Complex Role of STAT3 in Viral Infections. J. Immunol. Res. 2015, 2015, 272359. [Google Scholar] [CrossRef] [Green Version]
  57. Gao, G.; Luo, H. The ubiquitin-proteasome pathway in viral infections. Can. J. Physiol. Pharmacol. 2006, 84, 5–14. [Google Scholar] [CrossRef]
  58. Calistri, A.; Munegato, D.; Carli, I.; Parolin, C.; Palu, G. The ubiquitin-conjugating system: Multiple roles in viral replication and infection. Cells 2014, 3, 386–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Wang, P.; Zhao, W.; Zhao, K.; Zhang, L.; Gao, C. TRIM26 negatively regulates interferon-beta production and antiviral response through polyubiquitination and degradation of nuclear IRF3. PLoS Pathog. 2015, 11, e1004726. [Google Scholar] [CrossRef] [Green Version]
  60. Liu, J.; Huang, X.; Hao, S.; Wang, Y.; Liu, M.; Xu, J.; Zhang, X.; Yu, T.; Gan, S.; Dai, D.; et al. Peli1 negatively regulates noncanonical NF-kappaB signaling to restrain systemic lupus erythematosus. Nat. Commun. 2018, 9, 1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Slota, J.A.; Booth, S.A. MicroRNAs in Neuroinflammation: Implications in Disease Pathogenesis, Biomarker Discovery and Therapeutic Applications. Noncoding RNA 2019, 5, 35. [Google Scholar] [CrossRef] [Green Version]
  62. Li, J.; Song, Q.; Shao, L.; Zhang, L.L.; Guo, X.H.; Mao, Y.J. MiR-124a inhibits proliferation and invasion of rheumatoid arthritis synovial fibroblasts. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4581–4588. [Google Scholar] [PubMed]
  63. Tchankouo-Nguetcheu, S.; Bourguet, E.; Lenormand, P.; Rousselle, J.C.; Namane, A.; Choumet, V. Infection by chikungunya virus modulates the expression of several proteins in Aedes aegypti salivary glands. Parasit Vectors 2012, 5, 1–11. [Google Scholar] [CrossRef] [Green Version]
  64. Reshi, L.; Hong, J. Mitochondria as a Favourite Organelle for Invading Viruses. Mol. Biol. 2017, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  65. Maharaj, P.D.; Widen, S.G.; Huang, J.; Wood, T.G.; Thangamani, S. Discovery of mosquito saliva microRNAs during CHIKV infection. PLoS Negl. Trop. Dis. 2015, 9, e0003386. [Google Scholar] [CrossRef] [Green Version]
  66. Yen, P.S.; Chen, C.H.; Sreenu, V.; Kohl, A.; Failloux, A.B. Assessing the Potential Interactions between Cellular miRNA and Arboviral Genomic RNA in the Yellow Fever Mosquito, Aedes aegypti. Viruses 2019, 11, 540. [Google Scholar] [CrossRef] [Green Version]
  67. Goupil, B.A.; McNulty, M.A.; Martin, M.J.; McCracken, M.K.; Christofferson, R.C.; Mores, C.N. Novel Lesions of Bones and Joints Associated with Chikungunya Virus Infection in Two Mouse Models of Disease: New Insights into Disease Pathogenesis. PLoS ONE 2016, 11, e0155243. [Google Scholar] [CrossRef] [PubMed]
  68. Manimunda, S.P.; Vijayachari, P.; Uppoor, R.; Sugunan, A.P.; Singh, S.S.; Rai, S.K.; Sudeep, A.B.; Muruganandam, N.; Chaitanya, I.K.; Guruprasad, D.R. Clinical progression of chikungunya fever during acute and chronic arthritic stages and the changes in joint morphology as revealed by imaging. Trans. R. Soc. Trop. Med. Hyg. 2010, 104, 392–399. [Google Scholar] [CrossRef]
  69. Sims, N.A.; Gooi, J.H. Bone remodeling: Multiple cellular interactions required for coupling of bone formation and resorption. Semin. Cell Dev. Biol. 2008, 19, 444–451. [Google Scholar] [CrossRef]
  70. Friedenstein, A.J.; Petrakova, K.V.; Kurolesova, A.I.; Frolova, G.P. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968, 6, 230–247. [Google Scholar] [CrossRef]
  71. Parfitt, A.M. Osteonal and Hemi-Osteonal Remodeling: The Spatial and Temporal Framework for Signal Traffic in Adult Human Bone. J. Cell Biochem. 1994, 55, 273–286. [Google Scholar] [CrossRef]
  72. James, A.W. Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation. Scientifica 2013, 2013, 684736. [Google Scholar] [CrossRef] [PubMed]
  73. Shahi, M.; Peymani, A.; Sahmani, M. Regulation of bone metabolism. Rep. Biochem. Mol. Biol. 2017, 5, 73–82. [Google Scholar] [PubMed]
  74. Xu, J.; Li, Z.; Hou, Y.; Fang, W. Potential mechanisms underlying the Runx2 induced osteogenesis of bone marrow mesenchymal stem cells. Am. J. Transl. Res. 2015, 7, 2527–2535. [Google Scholar] [PubMed]
  75. Wang, C.; Liao, H.; Cao, Z. Role of Osterix and MicroRNAs in Bone Formation and Tooth Development. Med. Sci. Monit. 2016, 22, 2934–2942. [Google Scholar] [CrossRef] [Green Version]
  76. Komori, T. Regulation of osteoblast differentiation by transcription factors. J. Cell. Biochem. 2006, 99, 1233–1239. [Google Scholar] [CrossRef]
  77. Almalki, S.G.; Agrawal, D.K. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation 2016, 92, 41–51. [Google Scholar] [CrossRef] [Green Version]
  78. Hanna, H.; Mir, L.M.; Andre, F.M. In vitro osteoblastic differentiation of mesenchymal stem cells generates cell layers with distinct properties. Stem Cell Res. Ther. 2018, 9, 203. [Google Scholar] [CrossRef]
  79. Winslow, M.M.; Pan, M.; Starbuck, M.; Gallo, E.M.; Deng, L.; Karsenty, G.; Crabtree, G.R. Calcineurin/NFAT signaling in osteoblasts regulates bone mass. Dev. Cell. 2006, 10, 771–782. [Google Scholar] [CrossRef] [Green Version]
  80. Kim, K.; Lee, S.H.; Ha Kim, J.; Choi, Y.; Kim, N. NFATc1 induces osteoclast fusion via up-regulation of Atp6v0d2 and the dendritic cell-specific transmembrane protein (DC-STAMP). Mol. Endocrinol. 2008, 22, 176–185. [Google Scholar] [CrossRef] [Green Version]
  81. Macián, F.; García-Rodríguez, C.; Rao, A. Gene expression elicited by NFAT in the presence or absence of cooperative recruitment of Fos and Jun. EMBO J. 2000, 19, 4783–4795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Takayanagi, H.; Kim, S.; Koga, T.; Nishina, H.; Isshiki, M.; Yoshida, H.; Saiura, A.; Isobe, M.; Yokochi, T.; Inoue, J.; et al. Induction and Activation of the Transcription Factor NFATc1 (NFAT2) Integrate RANKL Signaling in Terminal Differentiation of Osteoclasts. Dev. Cell 2002, 3, 889–901. [Google Scholar] [CrossRef] [Green Version]
  83. Matsuo, K.; Galson, D.L.; Zhao, C.; Peng, L.; Laplace, C.; Wang, K.Z.; Bachler, M.A.; Amano, H.; Aburatani, H.; Ishikawa, H.; et al. Nuclear factor of activated T-cells (NFAT) rescues osteoclastogenesis in precursors lacking c-Fos. J. Biol. Chem. 2004, 279, 26475–26480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kim, K.; Kim, J.H.; Lee, J.; Jin, H.M.; Lee, S.H.; Fisher, D.E.; Kook, H.; Kim, K.K.; Choi, Y.; Kim, N. Nuclear factor of activated T cells c1 induces osteoclast-associated receptor gene expression during tumor necrosis factor-related activation-induced cytokine-mediated osteoclastogenesis. J. Biol. Chem. 2005, 280, 35209–35216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Matsumoto, M.; Kogawa, M.; Wada, S.; Takayanagi, H.; Tsujimoto, M.; Katayama, S.; Hisatake, K.; Nogi, Y. Essential role of p38 mitogen-activated protein kinase in cathepsin K gene expression during osteoclastogenesis through association of NFATc1 and PU.1. J. Biol. Chem. 2004, 279, 45969–45979. [Google Scholar] [CrossRef] [Green Version]
  86. Yasuda, H.; Shima, N.; Nakagawa, N.; Yamaguchi, K.; Kinosaki, M.; Mochizuki, S.; Tomoyasu, A.; Yano, K.; Goto, M.; Murakami, A.; et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc. Natl. Acad. Sci. USA 1998, 95, 3597–3602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lacey, D.L.; Timms, E.; Tan, H.L.; Kelley, M.J.; Dunstan, C.R.; Burgess, T.; Elliott, R.; Colombero, A.; Elliott, G.; Scully, S.; et al. Osteoprotegerin Ligand Is a Cytokine that Regulates Osteoclast Differentiation and Activation. Cell 1998, 93, 165–176. [Google Scholar] [CrossRef] [Green Version]
  88. Kim, K.; Kim, J.H.; Lee, J.; Jin, H.M.; Kook, H.; Kim, K.K.; Lee, S.Y.; Kim, N. MafB negatively regulates RANKL-mediated osteoclast differentiation. Blood 2007, 109, 3253–3259. [Google Scholar] [CrossRef]
  89. Miyauchi, Y.; Ninomiya, K.; Miyamoto, H.; Sakamoto, A.; Iwasaki, R.; Hoshi, H.; Miyamoto, K.; Hao, W.; Yoshida, S.; Morioka, H.; et al. The Blimp1-Bcl6 axis is critical to regulate osteoclast differentiation and bone homeostasis. J. Exp. Med. 2010, 207, 751–762. [Google Scholar] [CrossRef] [Green Version]
  90. Zhao, B.; Takami, M.; Yamada, A.; Wang, X.; Koga, T.; Hu, X.; Tamura, T.; Ozato, K.; Choi, Y.; Ivashkiv, L.B.; et al. Interferon regulatory factor-8 regulates bone metabolism by suppressing osteoclastogenesis. Nat. Med. 2009, 15, 1066–1071. [Google Scholar] [CrossRef] [Green Version]
  91. Kameda, T.; Mano, H.; Yuasa, T.; Mori, Y.; Miyazawa, K.; Shiokawa, M.; Nakamaru, Y.; Hiroi, E.; Hiura, K.; Kameda, A.; et al. Estrogen Inhibits Bone Resorption by Directly Inducing Apoptosis of the Bone-resorbing Osteoclasts. J. Exp. Med. 1997, 186, 489–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Jing, D.; Hao, J.; Shen, Y.; Tang, G.; Li, M.L.; Huang, S.H.; Zhao, Z.H. The role of microRNAs in bone remodeling. Int. J. Oral Sci. 2015, 7, 131–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Li, H.; Fan, J.; Fan, L.; Li, T.; Yang, Y.; Xu, H.; Deng, L.; Li, J.; Li, T.; Weng, X.; et al. MiRNA-10b Reciprocally Stimulates Osteogenesis and Inhibits Adipogenesis Partly through the TGF-beta/SMAD2 Signaling Pathway. Aging Dis. 2018, 9, 1058–1073. [Google Scholar] [CrossRef] [Green Version]
  94. Wei, F.; Yang, S.; Guo, Q.; Zhang, X.; Ren, D.; Lv, T.; Xu, X. MicroRNA-21 regulates Osteogenic Differentiation of Periodontal Ligament Stem Cells by targeting Smad5. Sci. Rep. 2017, 7, 16608. [Google Scholar] [CrossRef] [Green Version]
  95. Li, H.; Yang, F.; Wang, Z.; Fu, Q.; Liang, A. MicroRNA-21 promotes osteogenic differentiation by targeting small mothers against decapentaplegic 7. Mol. Med. Rep. 2015, 12, 1561–1567. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, S.; Wang, S.; Bian, C.; Yang, Z.; Zhou, H.; Zeng, Y.; Li, H.; Han, Q.; Zhao, R.C. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem. Cells Dev. 2012, 21, 2531–2540. [Google Scholar] [CrossRef] [Green Version]
  97. Kapinas, K.; Kessler, C.B.; Delany, A.M. miR-29 suppression of osteonectin in osteoblasts: Regulation during differentiation and by canonical Wnt signaling. J. Cell Biochem. 2009, 108, 216–224. [Google Scholar] [CrossRef] [Green Version]
  98. Wu, T.; Zhou, H.; Hong, Y.; Li, J.; Jiang, X.; Huang, H. miR-30 family members negatively regulate osteoblast differentiation. J. Biol. Chem. 2012, 287, 7503–7511. [Google Scholar] [CrossRef] [Green Version]
  99. Wang, J.; Guan, X.; Guo, F.; Zhou, J.; Chang, A.; Sun, B.; Cai, Y.; Ma, Z.; Dai, C.; Li, X.; et al. miR-30e reciprocally regulates the differentiation of adipocytes and osteoblasts by directly targeting low-density lipoprotein receptor-related protein 6. Cell Death Dis. 2013, 4, e845. [Google Scholar] [CrossRef] [Green Version]
  100. Chen, L.; Holmstrøm, K.; Qiu, W.; Ditzel, N.; Shi, K.; Hokland, L.; Kassem, M. MicroRNA-34a inhibits osteoblast differentiation and in vivo bone formation of human stromal stem cells. Stem Cells 2014, 32, 902–912. [Google Scholar] [CrossRef] [Green Version]
  101. Bae, Y.; Yang, T.; Zeng, H.C.; Campeau, P.M.; Chen, Y.; Bertin, T.; Dawson, B.C.; Munivez, E.; Tao, J.; Lee, B.H. miRNA-34c regulates Notch signaling during bone development. Hum. Mol. Genet. 2012, 21, 2991–3000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wei, J.; Shi, Y.; Zheng, L.; Zhou, B.; Inose, H.; Wang, J.; Guo, X.E.; Grosschedl, R.; Karsenty, G. miR-34s inhibit osteoblast proliferation and differentiation in the mouse by targeting SATB2. J. Cell Biol. 2012, 197, 509–521. [Google Scholar] [CrossRef] [Green Version]
  103. Shen, H.; Lu, C.; Shi, J.; Li, H.; Si, J.; Shen, G. Satb2 expression in Foxc1-promoted osteogenic differentiation of MC3T3-E1 cells is negatively regulated by microRNA-103–3p. Acta Biochim. Biophys. Sin. 2019, 51, 588–597. [Google Scholar] [CrossRef] [PubMed]
  104. Peng, H.; Lu, S.L.; Bai, Y.; Fang, X.; Huang, H.; Zhuang, X.Q. MiR-133a inhibits fracture healing via targeting RUNX2/BMP2. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2519–2526. [Google Scholar]
  105. Jia, J.; Tian, Q.; Ling, S.; Liu, Y.; Yang, S.; Shao, Z. miR-145 suppresses osteogenic differentiation by targeting Sp7. FEBS Lett. 2013, 587, 3027–3031. [Google Scholar] [CrossRef] [Green Version]
  106. Gu, Y.; Ma, L.; Song, L.; Li, X.; Chen, D.; Bai, X. miR-155 Inhibits Mouse Osteoblast Differentiation by Suppressing SMAD5 Expression. Biomed. Res. Int. 2017, 2017, 1893520. [Google Scholar] [CrossRef] [Green Version]
  107. Liu, H.; Zhong, L.; Yuan, T.; Chen, S.; Zhou, Y.; An, L.; Guo, Y.; Fan, M.; Li, Y.; Sun, Y.; et al. MicroRNA-155 inhibits the osteogenic differentiation of mesenchymal stem cells induced by BMP9 via downregulation of BMP signaling pathway. Int. J. Mol. Med. 2018, 41, 3379–3393. [Google Scholar] [CrossRef]
  108. Davis, C.; Dukes, A.; Drewry, M.; Helwa, I.; Johnson, M.H.; Isales, C.M.; Hill, W.D.; Liu, Y.; Shi, X.; Fulzele, S.; et al. MicroRNA-183–5p Increases with Age in Bone-Derived Extracellular Vesicles, Suppresses Bone Marrow Stromal (Stem) Cell Proliferation, and Induces Stem Cell Senescence. Tissue Eng. Part A 2017, 23, 1231–1240. [Google Scholar] [CrossRef]
  109. Huang, J.; Zhao, L.; Xing, L.; Chen, D. MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation. Stem Cells 2010, 28, 357–364. [Google Scholar] [CrossRef] [Green Version]
  110. Ito, K.; Tomoki, R.; Ogura, N.; Takahashi, K.; Eda, T.; Yamazaki, F.; Kato, Y.; Goss, A.; Kondoh, T. MicroRNA-204 regulates osteogenic induction in dental follicle cells. J. Dent. Sci. 2020. [Google Scholar] [CrossRef]
  111. Zheng, X.; Dai, J.; Zhang, H.; Ge, Z. MicroRNA-221 promotes cell proliferation, migration, and differentiation by regulation of ZFPM2 in osteoblasts. Braz. J. Med. Biol. Res. 2018, 51, e7574. [Google Scholar] [CrossRef] [PubMed]
  112. Gamez, B.; Rodriguez-Carballo, E.; Bartrons, R.; Rosa, J.L.; Ventura, F. MicroRNA-322 (miR-322) and its target protein Tob2 modulate Osterix (Osx) mRNA stability. J. Biol. Chem. 2013, 288, 14264–14275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tomé, M.; López-Romero, P.; Albo, C.; Sepúlveda, J.C.; Fernández-Gutiérrez, B.; Dopazo, A.; Bernad, A.; González, M.A. miR-335 orchestrates cell proliferation, migration and differentiation in human mesenchymal stem cells. Cell Death Differ. 2011, 18, 985–995. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, J.; Tu, Q.; Bonewald, L.F.; He, X.; Stein, G.; Lian, J.; Chen, J. Effects of miR-335–5p in modulating osteogenic differentiation by specifically downregulating Wnt antagonist DKK1. J. Bone Miner. Res. 2011, 26, 1953–1963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Long, H.; Zhu, Y.; Lin, Z.; Wan, J.; Cheng, L.; Zeng, M.; Tang, Y.; Zhao, R. miR-381 modulates human bone mesenchymal stromal cells (BMSCs) osteogenesis via suppressing Wnt signaling pathway during atrophic nonunion development. Cell Death Dis. 2019, 10, 470. [Google Scholar] [CrossRef] [Green Version]
  116. Kim, E.J.; Kang, I.H.; Lee, J.W.; Jang, W.G.; Koh, J.T. MiR-433 mediates ERRgamma-suppressed osteoblast differentiation via direct targeting to Runx2 mRNA in C3H10T1/2 cells. Life Sci. 2013, 92, 562–568. [Google Scholar] [CrossRef]
  117. Zhu, L.; Lin, Z.W.; Wang, G.; Zhang, H.; Liu, B.; Xu, Q.J. MicroRNA-495 downregulates AQP1 and facilitates proliferation and differentiation of osteoblasts in mice with tibial fracture through activation of p38 MAPK signaling pathway. Sci. Rep. 2019, 9, 16171. [Google Scholar] [CrossRef] [Green Version]
  118. Li, W.; Chen, Z.; Cai, C.; Li, G.; Wang, X.; Shi, Z. MicroRNA-505 is involved in the regulation of osteogenic differentiation of MC3T3-E1 cells partially by targeting RUNX2. J. Orthop. Surg. Res. 2020, 15, 143. [Google Scholar] [CrossRef] [Green Version]
  119. Zhang, J.F.; Fu, W.M.; He, M.L.; Wang, H.; Wang, W.M.; Yu, S.C.; Bian, X.W.; Zhou, J.; Lin, M.C.; Lu, G.; et al. MiR-637 maintains the balance between adipocytes and osteoblasts by directly targeting Osterix. Mol. Biol. Cell. 2011, 22, 3955–3961. [Google Scholar] [CrossRef] [Green Version]
  120. Hu, R.; Liu, W.; Li, H.; Yang, L.; Chen, C.; Xia, Z.Y.; Guo, L.J.; Xie, H.; Zhou, H.D.; Wu, X.P.; et al. A Runx2/miR-3960/miR-2861 regulatory feedback loop during mouse osteoblast differentiation. J. Biol. Chem. 2011, 286, 12328–12339. [Google Scholar] [CrossRef] [Green Version]
  121. Kim, K.; Kim, J.H.; Kim, I.; Lee, J.; Seong, S.; Park, Y.W.; Kim, N. MicroRNA-26a regulates RANKL-induced osteoclast formation. Mol. Cells 2015, 38, 75–80. [Google Scholar] [PubMed] [Green Version]
  122. Franceschetti, T.; Kessler, C.B.; Lee, S.K.; Delany, A.M. miR-29 promotes murine osteoclastogenesis by regulating osteoclast commitment and migration. J. Biol. Chem. 2013, 288, 33347–33360. [Google Scholar] [CrossRef] [Green Version]
  123. Lian, W.S.; Ko, J.Y.; Chen, Y.S.; Ke, H.J.; Hsieh, C.K.; Kuo, C.W.; Wang, S.Y.; Huang, B.W.; Tseng, J.G.; Wang, F.S. MicroRNA-29a represses osteoclast formation and protects against osteoporosis by regulating PCAF-mediated RANKL and CXCL12. Cell Death Dis. 2019, 10, 705. [Google Scholar] [CrossRef] [PubMed]
  124. Sul, O.J.; Rajasekaran, M.; Park, H.J.; Suh, J.H.; Choi, H.S. MicroRNA-29b Enhances Osteoclast Survival by Targeting BCL-2-Modifying Factor after Lipopolysaccharide Stimulation. Oxid. Med. Cell Longev. 2019, 2019, 6018180. [Google Scholar] [CrossRef] [PubMed]
  125. Mizoguchi, F.; Murakami, Y.; Saito, T.; Miyasaka, N.; Kohsaka, H. miR-31 controls osteoclast formation and bone resorption by targeting RhoA. Arthritis Res. Ther. 2013, 15, R102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Krzeszinski, J.Y.; Wei, W.; Huynh, H.; Jin, Z.; Wang, X.; Chang, T.C.; Xie, X.J.; He, L.; Mangala, L.S.; Lopez-Berestein, G.; et al. miR-34a blocks osteoporosis and bone metastasis by inhibiting osteoclastogenesis and Tgif2. Nature 2014, 512, 431–435. [Google Scholar] [CrossRef] [PubMed]
  127. Cong, F.; Wu, N.; Tian, X.; Fan, J.; Liu, J.; Song, T.; Fu, H. MicroRNA-34c promotes osteoclast differentiation through targeting LGR4. Gene 2017, 610, 1–8. [Google Scholar] [CrossRef]
  128. Lee, Y.; Kim, H.J.; Park, C.K.; Kim, Y.G.; Lee, H.J.; Kim, J.Y.; Kim, H.H. MicroRNA-124 regulates osteoclast differentiation. Bone 2013, 56, 383–389. [Google Scholar] [CrossRef]
  129. Tang, L.; Yin, Y.; Liu, J.; Li, Z.; Lu, X. MiR-124 Attenuates Osteoclastogenic Differentiation of Bone Marrow Monocytes Via Targeting Rab27a. Cell Physiol. Biochem. 2017, 43, 1663–1672. [Google Scholar] [CrossRef]
  130. Yang, S.; Zhang, W.; Cai, M.; Zhang, Y.; Jin, F.; Yan, S.; Baloch, Z.; Fang, Z.; Xue, S.; Tang, R.; et al. Suppression of Bone Resorption by miR-141 in Aged Rhesus Monkeys. J. Bone Miner. Res. 2018, 33, 1799–1812. [Google Scholar] [CrossRef] [Green Version]
  131. Wang, C.; He, H.; Wang, L.; Jiang, Y.; Xu, Y. Reduced miR-144–3p expression in serum and bone mediates osteoporosis pathogenesis by targeting RANK. Biochem. Cell Biol. 2018, 96, 627–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Li, N.; Liu, L.; Liu, Y.; Luo, S.; Song, Y.; Fang, B. miR-144–3p Suppresses Osteogenic Differentiation of BMSCs from Patients with Aplastic Anemia through Repression of TET2. Mol. Ther. Nucleic Acids. 2020, 19, 619–626. [Google Scholar] [CrossRef]
  133. Yu, F.Y.; Xie, C.Q.; Sun, J.T.; Peng, W.; Huang, X.W. Overexpressed miR-145 inhibits osteoclastogenesis in RANKL-induced bone marrow-derived macrophages and ovariectomized mice by regulation of Smad3. Life Sci. 2018, 202, 11–20. [Google Scholar] [CrossRef] [PubMed]
  134. Nakasa, T.; Shibuya, H.; Nagata, Y.; Niimoto, T.; Ochi, M. The inhibitory effect of microRNA-146a expression on bone destruction in collagen-induced arthritis. Arthritis Rheum. 2011, 63, 1582–1590. [Google Scholar] [CrossRef]
  135. Cheng, P.; Chen, C.; He, H.B.; Hu, R.; Zhou, H.D.; Xie, H.; Zhu, W.; Dai, R.C.; Wu, X.P.; Liao, E.Y.; et al. miR-148a regulates osteoclastogenesis by targeting V-maf musculoaponeurotic fibrosarcoma oncogene homolog B. J. Bone Miner Res. 2013, 28, 1180–1190. [Google Scholar] [CrossRef]
  136. Sul, O.J.; Sung, Y.B.; Rajasekaran, M.; Ke, K.; Yu, R.; Back, S.H.; Choi, H.S. MicroRNA-155 induces autophagy in osteoclasts by targeting transforming growth factor beta-activated kinase 1-binding protein 2 upon lipopolysaccharide stimulation. Bone 2018, 116, 279–289. [Google Scholar] [CrossRef]
  137. Zhao, C.; Sun, W.; Zhang, P.; Ling, S.; Li, Y.; Zhao, D.; Peng, J.; Wang, A.; Li, Q.; Song, J.; et al. miR-214 promotes osteoclastogenesis by targeting Pten/PI3k/Akt pathway. RNA Biol. 2015, 12, 343–353. [Google Scholar] [CrossRef]
  138. Liu, J.; Li, D.; Dang, L.; Liang, C.; Guo, B.; Lu, C.; He, X.; Cheung, H.Y.; He, B.; Liu, B.; et al. Osteoclastic miR-214 targets TRAF3 to contribute to osteolytic bone metastasis of breast cancer. Sci. Rep. 2017, 7, 40487. [Google Scholar] [CrossRef]
  139. Wang, W.; Yang, L.; Zhang, D.; Gao, C.; Wu, J.; Zhu, Y.; Zhang, H. MicroRNA-218 Negatively Regulates Osteoclastogenic Differentiation by Repressing the Nuclear Factor-kappaB Signaling Pathway and Targeting Tumor Necrosis Factor Receptor 1. Cell Physiol. Biochem. 2018, 48, 339–347. [Google Scholar] [CrossRef]
  140. Chen, C.; Cheng, P.; Xie, H.; Zhou, H.D.; Wu, X.P.; Liao, E.Y.; Luo, X.H. MiR-503 regulates osteoclastogenesis via targeting RANK. J. Bone Miner. Res. 2014, 29, 338–347. [Google Scholar] [CrossRef] [PubMed]
  141. Liu, T.; Qin, A.P.; Liao, B.; Shao, H.G.; Guo, L.J.; Xie, G.Q.; Yang, L.; Jiang, T.J. A novel microRNA regulates osteoclast differentiation via targeting protein inhibitor of activated STAT3 (PIAS3). Bone 2014, 67, 156–165. [Google Scholar] [CrossRef] [PubMed]
  142. Leong, K.-Y.B.; Chan, Y.-H.; Abdullah, W.M.A.N.W.; Lim, E.; Lai, K.-S. MicroRNAs in Bone Diseases: Progress and Prospects. In Transcriptional and Post-Transcriptional Regulation; IntechOpen: London, UK, 2018. [Google Scholar]
  143. Sun, Y.; Xu, L.; Huang, S.; Hou, Y.; Liu, Y.; Chan, K.M.; Pan, X.H.; Li, G. mir-21 overexpressing mesenchymal stem cells accelerate fracture healing in a rat closed femur fracture model. Biomed. Res. Int. 2015, 2015, 412327. [Google Scholar] [CrossRef] [PubMed]
  144. Salasznyk, R.M.; Klees, R.F.; Williams, W.A.; Boskey, A.; Plopper, G.E. Focal adhesion kinase signaling pathways regulate the osteogenic differentiation of human mesenchymal stem cells. Exp. Cell Res. 2007, 313, 22–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Eskildsen, T.; Taipaleenmäki, H.; Stenvang, J.; Abdallah, B.M.; Ditzel, N.; Nossent, A.Y.; Bak, M.; Kauppinen, S.; Kassem, M. MicroRNA-138 regulates osteogenic differentiation of human stromal (mesenchymal) stem cells in vivo. Proc. Natl. Acad. Sci. USA 2011, 108, 6139–6144. [Google Scholar] [CrossRef] [Green Version]
  146. Rodriguez-Carballo, E.; Gamez, B.; Ventura, F. p38 MAPK Signaling in Osteoblast Differentiation. Front. Cell Dev. Biol. 2016, 4, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Pathak, H.; Mohan, M.C.; Ravindran, V. Chikungunya arthritis. Clin. Med. 2019, 19, 381–385. [Google Scholar] [CrossRef]
  148. Birlik, M.; Koçak, A.; Harmanci, D. Role of MicroRNAs in Rheumatoid Arthritis. In New Developments in the Pathogenesis of Rheumatoid Arthritis; IntechOpen: London, UK, 2017. [Google Scholar]
  149. Ordas, A.; Kanwal, Z.; Lindenberg, V.; Rougeot, J.; Mink, M.; Spaink, H.P.; Meijer, A.H. MicroRNA-146 function in the innate immune transcriptome response of zebrafish embryos to Salmonella typhimurium infection. BMC Genom. 2013, 14, 696. [Google Scholar] [CrossRef] [Green Version]
  150. Lim, J.; Byun, J.; Guk, K.; Hwang, S.G.; Bae, P.K.; Jung, J.; Kang, T.; Lim, E.K. Highly Sensitive in Vitro Diagnostic System of Pandemic Influenza A (H1N1) Virus Infection with Specific MicroRNA as a Biomarker. ACS Omega 2019, 4, 14560–14568. [Google Scholar] [CrossRef]
  151. Janssen, H.L.; Reesink, H.W.; Lawitz, E.J.; Zeuzem, S.; Rodriguez-Torres, M.; Patel, K.; van der Meer, A.J.; Patick, A.K.; Chen, A.; Zhou, Y.; et al. Treatment of HCV infection by targeting microRNA. N. Engl. J. Med. 2013, 368, 1685–1694. [Google Scholar] [CrossRef] [Green Version]
  152. Stelma, F.; van der Ree, M.H.; Sinnige, M.J.; Brown, A.; Swadling, L.; de Vree, J.M.L.; Willemse, S.B.; van der Valk, M.; Grint, P.; Neben, S.; et al. A single dose of anti-miR-122, RG-101, in CHC patients results in NK cell normalization with no effect on HCV-specific CD8+ T cell function. Hepatology 2017, 66, 57–68. [Google Scholar] [CrossRef] [Green Version]
Figure 1. miRNAs regulate different signaling pathways in osteoblastogenesis.
Figure 1. miRNAs regulate different signaling pathways in osteoblastogenesis.
Viruses 12 01207 g001
Figure 2. miRNAs regulate different signaling pathways in osteoclastogenesis.
Figure 2. miRNAs regulate different signaling pathways in osteoclastogenesis.
Viruses 12 01207 g002
Table 1. miRNAs altered during Chikungunya virus (CHIKV) infection.
Table 1. miRNAs altered during Chikungunya virus (CHIKV) infection.
MicroRNAsTarget mRNAExpression Status
(Upregulated/
Downregulated)
References
aae-miR-2b3′ UTR of ubiquitin related modifier (URM), ubiquitin, and 3′ UTR of CHIKVUp[31]
hsa-miR-21-5pB-cell lymphoma 2 (BCL2), chemokine ligand 1 (CCL1), FASLG, pellino E3 ubiquitin protein ligase 1 (PELI1), interleukin 12 A (IL12A), transforming growth factor beta (TGFβ1)Up[25]
hsa-miR-138-2-3pTripartite motif containing 26 (TRIM5), TGF-Beta activated kinase 1 (MAP3K7) binding protein 3 (TAB3), tumor necrosis factor receptor superfamily (TNFRSF19), mitogen-activated protein kinase 13 (MAPK13), Apoptotic protease activating factor 1 (APAF1), Forkhead Box O3 (FOXO3)Up[25]
hsa-miR-146Tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6), interleukin-1 receptor-associated kinase 1 (IRAK1/2)Up[24]
hsa-miR-216a-5pCluster of Differentiation 6 (CD6), Janus kinase 2 (JAK2)Up[25]
aal-miR-305-3pECM receptor interaction, endocytosis, and SNARE interactions in vesicular transportUp[30]
hsa-miR-382-3pBeta-transducin repeat containing E3 ubiquitin protein ligase (BTRC), gap junction protein alpha 1 (GJA1), TRIM8, ubiquitin-conjugating enzyme E2 D2 (UBE2D2)Up[25]
hsa-miR-409-3pDNA topoisomerase 2-beta (TOP2B)Up[32]
hsa-miR-491-3pUBE2BUp[25]
hsa-miR-921nicotinic acid uptake protein (NIAP)Up[25]
aal-miR-927Soluble NSF attachment proteins receptor (SNARE) interactions in vesicular transportUp[30]
aae-miR-989sh2/sh3 adaptor and vacuolar ATP synthaseDown[31]
hsa-miR-1260aBCL2 antagonist/killer 1 (BAK1), activating transcription factor 6 beta (ATF6B)Up[25]
hsa-miR-1260bReceptor interacting serine/threonine kinase 1 (RIPK1), E3 ubiquitin-protein ligase NRDP1 (RNF41), suppressor of cytokine signaling 6 (SOCS6), NLR family CARD domain containing 5 (NLRC5), caspase 10 (CASP10)Up[25]
hsa-miR-1264TRIM26, bone morphogenetic protein 2 (BMP2), baculoviral IAP repeat containing 6 (BIRC6), interleukin 6 signal transducer (IL6ST), listerin E3 ubiquitin protein ligase 1 (LTN1), itchy E3 ubiquitin protein ligase (ITCH), MAPK8, SOCS5, UBE2D3Up[25]
hsa-miR-3074Integrin alpha-V (ITGAV), TNF receptor associated factor 3 (TRAF3)Up[25]
hsa-miR-4286Interferon alpha and beta receptor subunit 1 (IFNAR2), interleukin 13 receptor, alpha 1 (IL13RA1), interferon regulatory factor 1 (IRF1), ubiquitin conjugating enzyme E2 Z (UBE2Z), IL6RUp[25]
hsa-miR-4299SOCS7, signal transducer and activator of transcription 5 (STAT5B), TRIM28, mitogen-activated protein kinase kinase kinase 7 (MAP3K7), TAB1, AKT serine/threonine kinase 1 (AKT1), MAP3K11, MAP kinase-activated protein kinase 3 (MAPKAPK3), CAMP responsive element binding protein 1 (CREB1)Up[25]
hsa-miR-4443Interferon regulatory factor 3 (IRF3), mitogen-activated protein kinase kinase kinase 8 (MAP3K8), receptor interacting serine/threonine kinase 3 (RIPK3)Up[25]
hsa-miR-4695-3pCASP8, chemokine C-X-C motif ligand 2 (CXCL2)Up[25]
hsa-miR-4717-3pAKT3, UBE2M, sortilin (SORT1), ring finger protein 213 (RNF213), nerve growth factor receptor-associated protein 1 (NGFRAP1), MAPK10, IL11RA, C-C motif chemokine ligand 4 like 2 (CCL4L2), IFNAR1, IL7R, mitochondrial ubiquitin E3 ligase (MARCH5)Up[25]
hsa-miR-4762-5pNemo like kinase (NLK)Up[25]
hsa-miR-4775TNFRSF10A, toll-like receptors (TLR1), CASP3, ATF2, TAB2, glycogen synthase kinase 3 beta (GSK3B)Up[25]
hsa-miR-4794-5pUBE2S, STAT1, TRAF5, RIPK1, protein inhibitor of activated STAT 1 (PIAS1), AKT1Up[25]
hsa-miR-4878-3pDiablo IAP-Binding Mitochondrial Protein (DIABLO)Up[25]
hsa-miR-5100UBE2J1, axis inhibition protein 2 (AXIN2), IRAK4Up[25]
hsa-miR-5581-3pMAPK6, IL4R, MAP3K1, CCL18, death inducer-obliterator 1 (DIDO1)Up[25]
Table 2. The targets and effect of miRNAs on osteoblastogenesis.
Table 2. The targets and effect of miRNAs on osteoblastogenesis.
miRNATarget mRNAEffect on
Osteoblastogenesis
References
hsa-miR-10bSMAD2Enhances[93]
hsa-miR-21, mmu-miR-21SMAD5 (in human), SMAD7 (in mouse)Inhibits (in human), Enhances (in mouse)[94,95]
mmu-miR-22Histone deacetylase 6 (HDAC-6)Enhances[96]
mmu-miR-29ONInhibits[97]
mmu-miR-30RUNX2, SMAD1, LDL receptor related protein 6 (LRP6)Inhibits[98,99]
hsa-miR-34aNotch1, Notch2, and Jagged-1 (JAG1)Inhibits[100]
mmu-miR-34cNotch1, Notch2, and JAG1Inhibits[101]
mmu-miR-34sCYCLIN D1, CDK4, CDK6, special AT-rich sequence-binding protein 2 (SATB2)Inhibits[102]
mmu-miR-103-3pSATB2Inhibits[103]
hsa-miR-133RUNX2Inhibits[104]
hsa-miR-138Focal adhesion kinase (FAK)Inhibits
mmu-miR-145OSX gene (SP7)Inhibits[105]
mmu-miR-155SMAD5, RUNX2, and bone morphogenetic protein receptor type II (BMPR2)Inhibits[106,107]
mmu-miR-183Heme oxygenase 1 (HMOX-1)Inhibits[108]
mmu-miR-204 and hsa-miR-204RUNX2 (both in mouse and human), ALP (in human), ON (in human)Inhibits[109,110]
mmu-miR-221Zinc finger protein FOG family member 2 (ZFPM2)Enhances[111]
mmu-miR-322Transducer of ERBB2,2 (TOB2)Enhances[112]
hsa-miR-335RUNX2Inhibits[113]
mmu-miR-335-5pDickkopf WNT signaling pathway inhibitor 1 (DKK1)Enhances[114]
hsa-miR-381WNT5A, frizzled class receptor 3 (FZD3)Inhibits[115]
mmu-mir-433RUNX2Inhibits[116]
mmu-miR-495Aquaporin 1 (AQP1)Enhances[117]
mmu-miR-505RUNX2Inhibits[118]
hsa-miR-637OSXInhibits[119]
mmu-miR-2861HDAC-5 and homeobox A2 (HOXA2)Enhances[120]
mmu-miR-3960HDAC-5 and HOXA2Enhances[120]
Table 3. The targets and effect of miRNAs on osteoclastogenesis.
Table 3. The targets and effect of miRNAs on osteoclastogenesis.
miRNATarget mRNAEffect on OsteoclastogenesisReferences
mmu-miR-26aConnective tissue growth factor (CTGF)Inhibits[121]
mmu-miR-29 familyG protein-coupled receptor 85 (GPR85), CD93, nuclear factor I A (NFIA)Enhances[122]
mmu-miR-29aRANKL, CXCL12Inhibits[123]
mmu-miR-29bBcl-2-modifying factor (BMF)Enhances[124]
mmu-miR-31Rhodopsin (RHOA) and the GTPases of the RHO family (Ras-related C3 botulinum toxin substrate 1 (RAC1), RAC2, CDC42, RHOA, and RHOU)Inhibits[125]
mmu-miR-34aTGFB induced factor homeobox 2 (TGIF2)Inhibits[126]
mmu-miR-34cLeucine rich repeat containing G protein-coupled receptor 4 (LGR4)Enhances[127]
mmu-miR-124NFATC1, ras-related protein 27a (RAB27a)Inhibits[128,129]
mml-miR-141EPH receptor A2 (EPHA2)Inhibits[130]
hsa-miR-144-3pRANK, tet methylcytosine dioxygenase 2 (TET2)Inhibits[131,132]
mmu-miR-145SMAD3Inhibits[133]
mmu-miR-146aTRAF6 and IRAK-1Inhibits[134]
mmu-miR-148MAF BZIP transcription factor B (MAFB)Enhances[135]
mmu-miR-155TAB2Inhibits[136]
mmu-miR-214PTENEnhances[137]
mmu-miR-214-3pTRAF3Enhances[138]
mmu-miR-218TNFR1Inhibits[139]
hsa-miR-503RANKInhibits[140]
mmu-miR-9718PIAS3Enhances[141]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Roy, E.; Byrareddy, S.N.; Reid, S.P. Role of MicroRNAs in Bone Pathology during Chikungunya Virus Infection. Viruses 2020, 12, 1207. https://doi.org/10.3390/v12111207

AMA Style

Roy E, Byrareddy SN, Reid SP. Role of MicroRNAs in Bone Pathology during Chikungunya Virus Infection. Viruses. 2020; 12(11):1207. https://doi.org/10.3390/v12111207

Chicago/Turabian Style

Roy, Enakshi, Siddappa N. Byrareddy, and St Patrick Reid. 2020. "Role of MicroRNAs in Bone Pathology during Chikungunya Virus Infection" Viruses 12, no. 11: 1207. https://doi.org/10.3390/v12111207

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

Article Metrics

Back to TopTop