Archives of Oral Biology 72 (2016) 66–74
Contents lists available at ScienceDirect
Archives of Oral Biology journal homepage: www.elsevier.com/locate/aob
Review
The role of microRNA in periodontal tissue: A review of the literature Rizky Aditiya Irwandi, Anjalee Vacharaksa* Research Unit on Oral Microbiology and Immunology, Microbiology Department, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
A R T I C L E I N F O
Article history: Received 6 June 2016 Received in revised form 4 August 2016 Accepted 12 August 2016 Keywords: MicroRNA Bone remodeling Inflammatory mediators Periodontal tissue RANKL
A B S T R A C T
MicroRNAs (miRNAs) bind at the 30 UTR of their target mRNA to induce gene silencing. Through this mechanism, number of biological pathways implicated in developmental, physiological, and pathological processes, have been frequently found to involve miRNA functions. MiRNA functions in bone metabolism have also been reported, especially in association with receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclastogenesis. Expression of RANKL has been related to several inflammatory mediators, and thus some miRNAs may be implicated in the regulatory mechanism of inflammatoryinduced RANKL expression as shown in periodontal resident cells such as gingival fibroblasts or periodontal ligament cells. This review aims to review the current miRNA research relating periodontal tissue and its relevance in periodontal inflammation. In miRNA profiling studies of tissues isolated from individuals with periodontal disease, miR-223 has been consistently identified as a potential candidate miRNA to be further investigated in periodontitis-related processes. Although these studies point to an important role of miRNA-mediated epigenetic changes in tissue inflammation and alveolar bone loss, further investigation is still required to determine the function of miRNAs in the complex processes of periodontal tissue homeostasis and pathogenesis. Knowledge gained from future studies will be beneficial in developing alternative therapeutic approaches, especially ones that use miRNA delivery systems to treat periodontal disease. ã 2016 Elsevier Ltd. All rights reserved.
Contents 1. 2. 3. 4. 5.
MicroRNA biology and function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Bone remodeling and the role of inflammatory mediators in periodontal tissue . The role of miRNA in bone remodeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . MicroRNA dynamics in periodontal tissue and periodontitis-related mechanisms Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1. MicroRNA biology and function Epigenetic regulation plays a role in defining the specificity of gene expression in living organisms. Approximately 0.5% of the human genome varies from person to person. Chimpanzees and humans, on the other hand, share 98% of their genomes, yet are very distinct from one another (Varki & Altheide, 2005). The phenotypic differences within and between species with very similar genomes probably result largely from epigenetic control.
* Corresponding author. E-mail address:
[email protected] (A. Vacharaksa). http://dx.doi.org/10.1016/j.archoralbio.2016.08.014 0003-9969/ã 2016 Elsevier Ltd. All rights reserved.
.... .... .... ... .... ....
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
66 67 68 70 72 72
Each cell that makes up an organism has the ability to alter chromatin structure, which results in changes in DNA sequence read-out. Epigenetic mechanisms, including DNA methylation, histone modification, chromatin remodeling, and small interfering RNAs, are integrated in cooperative and regulatory networks to control biological changes (Murr, 2009). Studies on small RNAs, especially microRNAs (miRNAs), have been increasing substantially over the past two decades, since the discovery of miRNA in 1993 (Lee, Feinbaum, & Ambros, 1993; Almeida, Reis, & Calin, 2011). A number of biological pathways, implicated in developmental, physiological, and pathological processes, have been frequently found to involve miRNA functions (Neo et al., 2014; Lin, Yue, Pan, Sun, & Wang, 2011; Parikh et al., 2014).
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
The miRNA is composed of approximately 22 nucleotides. Six of these nucleotides at the 50 end, known as ‘seed sequence’, can specifically bind to 30 UTRs of target messenger RNAs (mRNAs), leading to translational repression (Huntzinger & Izaurralde, 2011; Pasquinelli, 2012). The miRNA can cause structural disruption of mRNAs in two different ways, depending on the number of basepairing with its target mRNA. For translational blockage in plant cells, the 22-nucleotide miRNA is required to completely bind to its target mRNA (Brodersen et al., 2008), leading to the degradation of the target mRNA through the endonuclease activity of the miRNA silencing machinery. In contrast, partial pairing of the 6-nucleotide of the seed sequence is adequate to induce the de-adenylation of the target mRNA resulted in structural disruption (Pasquinelli, 2012) and translational blockage in animal cells (Bartel, 2009). In the canonical targeting pattern, 6-mer at positions 2–8 of miRNA is perfectly matched with the sequence of the transcript while the imperfect match refers to a non-canonical type (Hausser & Zavolan, 2014). The canonical type effectively causes target repression (Wang, 2014). By these mechanisms, each miRNA, not only directly represses a number of target genes, but also indirectly affects other associated genes to regulate cell functions (Baek et al., 2008; Selbach et al., 2008). Although all mechanisms can block translation, whether silencing occurs predominantly by mRNA structural disruption or by translational interference remains controversial. MiRNA biogenesis is the process by which pri-miRNAs are sequentially cleaved to first generate pre-miRNAs, which are in turn cleaved to produce the mature miRNAs. The process starts in the nucleus, where pri-miRNAs are transcribed by the RNA polymerase II. The pri-miRNA is a long primary transcript with a local stem-loop structure that contains a local hairpin structure encoding the miRNA sequence. The microprocessor complex, composed of Drosha and DiGeorge Syndrome Chromosomal Region 8 (DGCR8), subsequently cleaves the pri-miRNA to produce the pre-miRNA (Han et al., 2004). The pre-miRNA is then exported from the nucleus to the cytoplasm by exportin-5. In the cytoplasm, pre-miRNA is cleaved to become an 18–25-nucleotide-long double-stranded RNA (dsRNA) through the function of Dicer, an RNase III-type endonuclease that acts specifically on dsRNAs. One strand of this hairpin duplex is loaded into the Argonaute (AGO) protein to form the RNA-induced silencing complex (RISC). The mature miRNAs in complex with RISC are able to target mRNAs through base-pairing (Ha & Kim, 2014). MiRNAs play various roles in complex biological cascades including the inflammatory responses. The shift in miRNA profiles in the gingiva and salivary gland, for example, indicates the miRNA-related host immune response to oral bacterial infection at the primary and secondary infection sites in a rat model (Nayar et al., 2016). In autoimmune disease, increase of miR-148a enhances B cells auto-reactivity by targeting the autoimmune suppressor, Gadd45a, the tumor suppressor, PTEN and proapoptotic protein, Bim (Gonzalez-Martin et al., 2016), while anti-inflammatory effect of miR-24 is reported in macrophage (Fordham, Naqvi, & Nares, 2015). The inflammatory mediator, tumor necrosis factor (TNF)-a, activates osteoclastogenesis in mouse bone marrow-derived precursor cells through the upregulation of miR-182 (Miller et al., 2016). Thus, miR-182 inhibitor may represent an effective regulator to control inflammatory osteoclastogenesis and bone resorption. These findings emphasize the important functions of miRNAs and epigenetic regulation. This review will discuss the role of miRNAs in healthy and inflamed periodontal tissues as well as the miRNAs in inflammatory-related bone remodeling that have been reported in periodontal resident cells. The dynamics of miRNA profiles in periodontal tissue as related to periodontitis is summarized.
67
2. Bone remodeling and the role of inflammatory mediators in periodontal tissue The physiologic bone remodeling around teeth or around dental implants is similar at the cellular level to bone remodeling that occurs in other bone tissues in the human body. Osteocytes, osteoclasts, and osteoblasts function to balance the processes of bone resorption and bone formation. The bone remodeling process is initiated by several factors, including parathyroid hormones (Shah et al., 2004), mechanical load (Yang et al., 2004), growth factors, and cytokines (Raggatt & Partridge, 2010), which result in the reorganization of the cellular and extracellular matrices (Ståhle-Bäckdahl et al., 1997) and changes in local vascularity (Matsuzaki, Wohl, Novack, Lynch, & Silva, 2007). This bone remodeling process leads to the synthesis and release of various neurotransmitters (Grässel, 2014), arachidonic acid (Dean et al., 2008), growth factors, metabolites, cytokines (Mundy, 1993), colony-stimulating factors (Raisz, 1999), and enzymes like cathepsin K (Crockett, Rogers, Coxon, Hocking, & Helfrich, 2011), matrix metalloproteinases (MMP) (Sasaki et al., 2007), and aspartate aminotransferase (Perinetti et al., 2003). During remodeling, osteoblasts mediate collagen degradation through the release of MMP-13, which results in the exposure of an arginyl-glycyl-aspartic acid motif for osteoclast binding (McHugh et al., 2000). Osteoblasts also release monocyte chemoattractant protein-1 (MCP-1) to recruit pre-osteoclasts. In addition, they express the receptor activator of nuclear factor kappa B ligand (RANKL), which then binds to its receptor, RANK, expressed on osteoclast precursor cells (Udagawa et al., 1999). The binding of RANK and RANKL directs the fate of hematopoietic progenitor cells to the osteoclast lineage and is a distinctive mark of osteoclast development. Indeed, conditional RANK (Dougall et al., 1999) or RANKL (Kong et al., 1999) knockout mice show no osteoclast differentiation. RANK-RANKL interaction is inhibited by the competitive binding of osteoprotegerin (OPG) to RANKL, which leads to the suppression of osteoclast differentiation (Wada, Nakashima, Hiroshi, & Penninger, 2006). Because of their important roles in osteoclastogenesis, RANK, RANKL, and OPG are tightly regulated during this process. Inflammatory bone remodeling through RANK/RANKL/OPG interaction is summarized in Fig. 1. In inflamed periodontal tissue, prostaglandin E2 (PGE2) and other inflammatory cytokines induce RANKL expression to promote osteoclastogenesis. PGE2 is the product of arachidonic acid conversion by cyclooxygenase-2 (COX-2). In periodontal tissue, Porphyromonas gingivalis lipopolysaccharide (PgLPS) stimulates PGE2 release (Noguchi et al., 1996) and increases the RANKL/ OPG ratio in primary human gingival fibroblasts (Belibasakis et al., 2007). The receptors for PGE2, E prostanoid (EP) 2 and EP4, play a role in PGE2-induced bone resorption (Minamizaki, Yoshiko, Kozai, Aubin, & Maeda, 2009). After PGE2 treatment, RANKL expression is increased in mouse calvarial osteoblasts. In addition to PGE2, the interleukin (IL)-6 cytokine may also play a role in osteoclastogenesis. IL-6 is produced by leukocytes, macrophages, periodontal ligament cells, and gingival fibroblasts in the periodontal tissue (Nebel, Arvidsson, Lillqvist, Holm, & Nilsson, 2013), and its levels is increased in response to IL-1, tumor necrosis factor a (TNFa), viruses, bacterial toxins, or lipopolysaccharide (LPS) (Morandini et al., 2010). Although IL-6 has both pro- and anti-inflammatory properties (Scheller, Chalaris, Schmidt-Arras, & Rose-John, 2011), it has been shown to increase RANKL and PGE2 expression in mouse calvarial osteoblasts (Palmqvist, Persson, Conaway, & Lerner, 2002) and in fibroblast-like synoviocytes of rheumatoid arthritis patients (Hashizume, Hayakawa, & Mihara, 2008) through the activation of the JAK/STAT signalling pathway. Co-stimulation of IL-6 and PGE2 leads to higher expression of COX-2, EP2, and EP4 and to enhanced
68
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
Fig. 1. The interaction of RANK/RANKL/OPG in inflamed periodontal tissue. Periodontal pathogens stimulate the inflammatory cytokine release, including TNF-a, interleukin (IL)-1b, IL-6, and prostaglandin E2 (PGE2), in association with periodontitis. The inflammatory cytokines enhance the differentiation of osteoclast precursors and induce RANKL expression in gingival fibroblast, periodontal ligament cells and osteoblasts to promote osteoclastogenesis. Osteoblasts express the receptor activator of nuclear factor kappa B ligand (RANKL), which then binds to its receptor, RANK, expressed on osteoclast precursor cells. While osteoprotegerin (OPG) can competitively bind to RANKL and suppress osteoclast differentiation, the interaction of RANK and RANKL directs the fate of hematopoietic progenitor cells to the osteoclast lineage and is a distinctive mark of osteoclast development.
bone resorption in mouse calvarial osteoblasts (Liu, Kirschenbaum, Yao, & Levine, 2006). The pro-inflammatory cytokines, TNF-a (Singh, Gupta, Bey, & Khan, 2014) and IL-1b (Sánchez, Miozza, Delgado, & Busch, 2013), are also highly associated with the initiation of periodontitis. IL-1b enhances the differentiation of osteoclast precursors and induces RANKL expression in several cell types by promoting the interactions between interleukin (IL)-1, IL-1 receptor type 1 (IL-1R1), and IL-1 receptor accessory protein (IL-1RAcp), which activate the ERK and p38 MAPK signalling pathways (Steeve, Marc, Sandrine, Dominique, & Yannick, 2004; Mine, Makihira, Yamaguchi, Tanaka, & Nikawa, 2014). The activation of these pathways results in the increase of RANKL expression in primary human periodontal ligament cells (Kanzaki, Chiba, Shimizu, & Mitani, 2002; Nukaga et al., 2004), osteoblasts (Choi, Moon, Cha, Kim, & Yoo, 2005), gingival fibroblasts, and periodontal ligament fibroblasts (Belibasakis et al., 2007; Nukaga et al., 2004). TNFa also enhances RANKL-induced osteoclastogenesis via the coupling of TNFR1 and RANK signalling pathways (Zhang, Heulsmann, Tondravi, Mukherjee, & Abu-Amer, 2001). Pathogens, as well as endotoxins and related substances, can stimulate TNFa release from monocytes, macrophages, or T cells (Kitaura et al., 2013). Interestingly, recombinant RANKL also increases TNFa expression in mouse bone marrow (Zou, Hakim, Tschoep, Endres, & Bar-Shavit, 2001), where TNFa then stimulates the differentiation of macrophages into osteoclasts, as demonstrated by TRAP-positive staining. This process has been shown to be mediated by TNFa receptor type 1 (TNFR1) and type 2 (TNFR2) in vitro (Kobayashi et al., 2000) and in vivo (Lam et al., 2000). 3. The role of miRNA in bone remodeling In bone metabolism, some miRNAs are involved as stimulatory or inhibitory factors of osteogenesis. miRNA-194 stimulates osteoblast differentiation by targeting chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) in vitro (Jeong,
Kang, Hwang, Kim, & Koh, 2014). In mouse mesenchymal stem cells cultured in osteogenic medium, miR-194 is upregulated, while COUP-TFII is suppressed, by high levels of runt-related transcription factor 2 (RUNX2) (Lee et al., 2012). This result is reversed by the addition of anti-miR-194 (Jeong et al., 2014). miR-542-3p, on the other hand, inhibits osteoblast differentiation and proliferation by targeting BMP-7 and decreasing RUNX2-, type 1 collagen-, osterix-, and osteocalcin-specific mRNAs in mouse calvarial osteoblasts. miR-542-3p also inhibits bone formation and decreases the rate of mineral apposition in wild type and ovariectomized Balb/c mice (Kureel et al., 2014). MiRNAs also play a role in osteoclastogenesis. Protein inhibitor of activated STAT 3 (PIAS3) is a negative regulator for osteoclastogenesis by inhibiting transcriptional activity of microphthalmiaassociated transcription factor (MITF) (Kim et al., 2007; Hikata et al., 2009) and miR-9718 targets PIAS3 during osteoclast differentiation (Liu et al., 2014). Transfection of pre-miR-9718 into RAW 267.7 cells leads to lower expression of PIAS3, but higher expression of osteoclastogenic markers such as nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1 (NFATc1), c-Fos, MITF, and nuclear factor kappa B (NFkB). A higher number of TRAP-positive cells after pre-miR-9718 transfection also indicates an increase in osteoclastogenesis. Conversely, intravenous injection of antagomir-9718 in both wild-type and ovariectomized mice leads to restored bone mineral density (Liu et al., 2014). In contrast to miR-9718, miR-34a plays a role in the inhibition of osteoclastogenesis. In an in vivo model, miR-34a has been shown to target TGIF2 (Krzeszinski et al., 2014), which leads to the repression of TGFb-responsive genes (Melhuish, Gallo, & Wotton, 2001) and the inhibition of osteoclast differentiation (Quinn et al., 2001). miR-34a knockout mice exhibit decreased TRAP mRNA and TRAP-positive cell number, but increased bone quantity, when compared to wild type mice (Krzeszinski et al., 2014). miR-34a is significantly downregulated after an increase in RANKL or after treatment with rosiglitazone, a drug to induce bone loss. Further analysis shows that transfection of synthetic pre-miR-34a mimics into human
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
peripheral mononuclear cells (PBMC) led to decreased TRAPspecific mRNA and TRAP-positive cell number. In contrast, transfection of an inhibitor of pre-miR-34a resulted in increased TRAP-specific mRNA and TRAP-positive cell number indicating miR-34a suppressed osteoclastogenesis in vitro (Krzeszinski et al., 2014). Delivery of miR-34a using chitosan nanoparticles to the ovariectomized and two-cancer-cell-cardiac-injection mouse models results in higher bone quantity and lower bone metastatic rate compared with control ovariectomized mice (Krzeszinski et al., 2014). Several miRNAs have been reported to be involved in the regulation of RANKL expression in various cell types, as summarized in Table 1. In cancer research, miR-335 has been shown to inhibit small cell lung cancer (SCLC) bone metastases by targeting insulin-like growth factor-1 receptor (IGF1R) and RANKL (Gong et al., 2014). Xenografts of the SCLC SBC-5, but not SBC-3, cell line triggers the formation of osteolytic bone lesions in non-obese diabetic/severe-combined and immune-deficient (NOD/SCID) IL2Rg null mice that lack mature T cells, B cells, and functional NK cells (Liu et al., 2014). A microarray study using the mParaflo1Microfluidic Biochip Microarray shows that 14 out of 833 miRNAs are downregulated in SBC-5 mice when compared with SBC-3 mice. This phenotype is thought to arise from the lack of miR-335 in SBC-5. Consistent with this, SBC-5 mice transfected with miR-355 show lower levels of both IGF1R and RANKL protein expression and lower incidence of bone lesions (Gong et al., 2014). The level of RANKL expression appears to be associated with osteoclastogenesis. In giant cell tumour of bone, miR-106b has been shown to inhibit osteoclastogenesis and osteolysis by directly targeting RANKL. Transfection of miR-106b into both giant cell tumour stromal cells and MG63 osteosarcoma cells directly targets RANKL mRNA, as shown by luciferase reporter assay, and reduces osteoclastogenesis in vivo (Wang et al., 2015). In contrast, glucocorticoid treatment induces secondary osteoporosis with bone loss and fragility fracture (Fraser & Adachi, 2009). In MC3T3E1 cells and an in vivo model, glucocorticoid treatment increases the level of RANKL while decreasing the level of miR-17/20a (Shi
69
et al., 2014). The bone loss associated with glucocorticoid treatment seems to be partly rescued by the addition of miR-17/ 20a (Shi et al., 2014). These observations point to the role of miRNAs in RANKL regulation, which in turn affects bone metabolism. MiRNAs play a role in inflammatory mediator-induced osteoclastogenesis by targeting genes that encode inflammatory mediators or by ultimately affecting the release of these mediators and we summarized the relevant studies of it in Table 2. Rheumatoid arthritis shows chronic inflammatory characteristics similar to periodontitis (Agnihotri & Gaur, 2015). The presence of TNFa and IL-1b in rheumatoid arthritis can induce miR-146a expression in synovial fibroblasts (Nakasa et al., 2008). The NFkBdependent induction of miR-146a targets TRAF6 and IRAK1 (Taganov, Boldin, Chang, & Baltimore, 2006), which leads to the inhibition of TNFa, IL-1b, and IL-6 in human gingival fibroblasts (Xie et al., 2013). IL10-induced miR-187, on the other hand, decreases IL-6 production in primary human monocytes in vitro (Rossato et al., 2012). The consequences of miRNA modulation that affects inflammatory mediator release can be inducer-dependent and cell type-dependent. For instance, bleomycin-induced senescent HCA2 cells express higher miR-146a compared with untreated cells, resulting in reduced IL-6 and IL-8 production through IRAK1 inhibition (Bhaumik et al., 2009). PgLPS-induced miR-146 expression in human primary gingival fibroblasts, on the other hand, suppresses not only IL-6 but also IL-1b and TNFa via IRAK1 inhibition (Xie et al., 2013). Moreover, miRNA can also be induced by cytokines affecting other cytokine release. For example, IL-10induced miR-187, which suppressed TNFa and IL-6 through targeting TNFa-specific and NFkB inhibitor zeta-specific mRNA, respectively in human primary monocytes (Rossato et al., 2012). The effects of constitutive miRNA expression can vary between cell types. Fibroblast-like synoviocytes from rheumatoid arthritis patients express miR-146a, which reduces LPS-induced TNFa production by destabilizing TNFa-specific mRNA (Semaan et al., 2011). In SJL mice-derived bone marrow stem cells, on the other hand, miR-146a reduces PGE2 production by targeting PGE2
Table 1 The miRNAs thatare involved in bone metabolism and RANKL expression. miRNAs
Functions
Cell/tissue types
Reference
miR-194
Stimulation of osteogenesis through inhibition of COUP-TFII, RUNX2 suppressor protein.
McHugh et al. (2000)
miR-5423p
Inhibition of osteogenesis through suppression of bone morphogenetic protein (BMP-7)
miR-9718
Stimulation of osteoclastogenesis through inhibition of PIAS3
miR-34a
Inhibition of osteoclastogenesis through suppression of transforming growth factor ß-induced factor II (TGIFII)
miR-335
Inhibition of small cell lung cancer bone metastases by suppression of IGF1R and RANKL expression
miR-17 miR20a
Inhibition of glucocorticoid-induced osteoclastogenesis by suppressing RANKL expression
miR-106b
Inhibition of osteoclastogenesis and osteolysis through RANKL repression
primary mouse bone marrow stromal cells MC3T3-E1 cells In vitro Mouse calvarial osteoblasts In vitro Balb/c mice In vivo C57BL/6-derived bone marrow cells In vitro Wild type and ovariectomized C57BL/6 mice In vivo C57BL/6J-derived bone marrow cells In vitro miR-34a transgenic C57BL/6J mice In vivo SBC-5: small cell lung cancer cell line In vitro C57BL/6J mice In vivo Mouse calvarial osteoblasts: primary cells, MC3T3-E1 cell line, calvarial bone In vitro C57BL/6J mice In vivo Giant cell tumour In vitro Wild type and ovariectomized C57BL/6 mice In vivo
Miller et al. (2016)
Moffatt and Lamont (2011)
Morandini et al. (2010)
Murr (2009)
Nebel et al. (2013)
Nakasa et al. (2008)
70
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
Table 2 The role of miRNAs in the release of inflammatory mediator inducing RANKL expression. miRNAs
Functions
Inducer
Cell/tissue type
Reference
miR-146
Suppression of IL-6 & IL-8 by targeting IRAK1
Senescent induction P. gingivalis LPS –
Human foreskin fibroblasts cell line, HCA2 In vitro Human primary gingival fibroblasts In vitro Human primary rheumatoid arthritis fibroblast-like synoviocytes In vitro Human primary monocytes In vitro Human primary chondrocytes In vitro Human primary umbilical monocyte In vitro SJL mice-derived bone marrow stem cells In vitro
Palmqvist et al. (2002) Ogata et al. (2014) Parikh et al. (2014)
Suppression of IL-6, IL-1b, and TNFa by targeting IRAK1 miR-346
Suppression of LPS-activated TNFa production by tristetrapolin stabilization of TNFa-specific mRNA
miR-187
IL-10
miR-199
Suppression of TNFa by targeting TNFa-specific mRNA Suppression of IL-6 by targeting NFkB inhibitory zeta Suppression of PGE2 synthesis by targeting COX-2 mRNA
miR-125b
Suppression of constitutive or LPS-stimulated TNFa production
–
miR-146a
Inhibition of PGE2 synthesis by targeting PGE2 synthase 2 mRNA
–
synthase 2-specific mRNA (Matysiak et al., 2013). In human primary chondrocyte, miR-199 also reduces PGE2 production by targeting COX-2-specific mRNA (Akhtar & Haqqi, 2012). However, constitutive miRNA expression can also modulate cytokine release constitutively and inductively, such as when miR-125b suppresses both constitutive and LPS-induced TNFa production in human primary umbilical monocytes (Huang et al., 2012). 4. MicroRNA dynamics in periodontal tissue and periodontitisrelated mechanisms The role of miRNAs in periodontal disease remains to be elucidated. Recent studies have investigated the profile of miRNAs in inflamed gingiva in comparison with healthy tissue summarized in Table 3. Two studies using the miRNA PCR array (SABioscience) reveal that several miRNAs are increased in inflamed periodontal tissue, including miR-181b, miR-19b, miR-23a, miR-30a, miR-let7a, and miR-301a, miR-128, miR-34a, and miR-381 (Lee et al., 2011; Na et al., 2016). Some miRNAs, including miR-211, miR-372, miR-656, are underexpressed in inflamed gingival tissue compared with healthy gingiva (Na et al., 2016). A study using the miRCURYTM array reports a greater than 2-fold increase in 91 miRNAs, and a greater than 2-fold decrease in 34 miRNAs in inflamed gingival tissue. Twelve of the 91 up-regulated miRNAs, including miR-1265p, miR-20a, miR-142-3p, miR-19a, let-7f, miR-203, miR-17, miR-223, miR-146a, miR-146b, miR-155, and miR-205, are related to inflammation, as predicted by TargetScan and the database on miRNA.org (Xie, Shu, Jiang, Liu, & Zhang, 2011) that contains miRNA target prediction based on miRANDA algorithm (Betel, Wilson, Gabow, Marks, & Sander, 2008). Another study using the Agilent Human miRNA Microarray shows that miR-150, miR-223, and miR-200b have the highest miRNA expression, while miR-199a5p and miR-214 had the lowest miRNA expression in inflamed tissue (Ogata et al., 2014). Interestingly, miR-150, miR-223, and miR-200b are predicted by Ingenuity Pathway Analysis (IPA) to associate with inflammatory disease, tissue injury, leukocytic and hormonal abnormalities, urological disease, and cancer (Ogata et al., 2014). A large-scale genome-wide microarray study (Stoecklin-Wasmer et al., 2012) that assessed over 1000 miRNAs in 198 gingival tissues shows that the expression of 159 miRNAs are significantly different when compared between healthy and inflamed gingiva. Four miRNAs, including miR-451, miR-223, miR-486-5p, and miR-3917, are significantly overexpressed, whereas seven miRNAs, including miR-1246, miR-1260, miR-141, miR1260b, miR-203, miR-210, and miR-205-3p, are under-expressed in inflamed gingiva. The five studies discussed above reveal distinct miRNA profiles for inflamed gingival tissues. This may be due to age and gender
–
Ouhara et al. (2014) Perinetti et al. (2003) Perri et al. (2012) Pasquinelli (2012)
variations among the study samples, differences in criteria for gingival inflammation, variable environmental exposure of individuals, especially at the tissue collection site, and differences in probes used for the microarrays. The inclusion of complex cells from the gingival tissue samples may also have contributed to the variability of miRNA expression patterns. Interestingly, however, three microarray studies consistently report the up-regulation of miR-223 in inflamed gingiva. miR-223 targets nuclear factor 1-A (NF1-A), and down-regulation of miR-223 by antisense transfection into RAW264.7 cells or by lentivirus-mediated silencing in a mouse model (Lee et al., 2011) has been found to reduce osteoclast formation by up-regulating NF1-A. PU.1, a transcription factor that defines osteoclast function, has also been found to enhance osteoclast formation by up-regulating miR-223 (Sugatani & Hruska, 2009). These studies suggest that miR-223 levels in inflamed gingival tissue may play a role in alveolar bone loss, which is a hallmark of periodontitis. The role of miRNAs in host immune response and inflammation through the regulation of inflammatory cytokines has been proposed in recent studies. Interestingly, based on miRNA profiling of overweight patients with periodontitis, obesity is thought to be a risk factor in periodontal tissue inflammation (Perri, Nares, Zhang, Barros, & Offenbacher, 2012). Inflammation-related miRNAs, including miR-18a and miR-30e, have been shown to be increased in obese patients, compared with non-obese patients, with healthy periodontium. While only miR-30e and miR-106b are significantly overexpressed in non-obese patient with periodontitis, miR-15a, miR-18a, miR-22, miR-30d, miR-30e, miR-103, miR106b, miR-130a, miR-142-3p, miR-185, and miR-210 are significantly overexpressed in obese patients with periodontitis. Functional analyses of target mRNAs using miRNA bioinformatic analysis and Gene Ontology (GO) shows that the miRNAs identified may be involved in the regulatory pathways of immunity and inflammation and the metabolism of collagen, lipid, and carbohydrate (Perri et al., 2012). A more recent study (Kalea et al., 2015) also reports the association of miR-200b with obesity and periodontitis, and its targeted genes are involved in wound healing and angiogenesis. Because of the small sample size of these studies, the role of miRNAs in regulating periodontitis and the impact of obesity in disease pathogenesis remain inconclusive. However, some miRNAs with increased expression in the group of obese patients with periodontitis may be significant. Among these upregulated miRNAs, miR-106b has been linked to carcinogenesis, cell-cycle regulation, inflammation, and bone metabolism. The detection of some miRNAs as a biomarker for periodontitis is also discussed. The use of saliva or gingival crevicular fluid as a diagnostic medium for detection of miRNA biomarkers is noninvasive and accessible (Xie et al., 2015). From the miRNAs that
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
71
Table 3 Summary of recent reports in the profile of miRNA in inflamed gingiva compared with healthy gingival tissue. Author
Lee et al., 2011
Xie et al., 2012
StoecklinWasmer et al., 2012
Ogata et al., 2014
Na et al., 2016
Sample
Array
miRNAs
Criteria
Number
Age/ Gender
Inflamed gingiva: Chronic periodontitis PD > 5 mm; AL > 3 mm; Bone loss on radiograph Healthy gingiva: Low scores on BoP (under 10%); No site with PD > 3 mm No site presenting AL Inflamed gingiva: GI > 1; PD 5 mm; AL 3 mm at least 5 sites; Bone loss on radiograph Healthy gingiva: PD > 3 mm; AL < 1 mm; No bone loss on radiograph Inflamed gingiva: PD > 4 mm; AL 3 mm; BoP Healthy gingiva: PD 4 mm; AL 2 mm.; no BoP Inflamed gingiva: PD 6 mm AL > 6 mm Healthy gingiva: Edentulous residual ridges Inflammed gingiva: PD > 5 mm; AL > 3 mm; no bone loss on radiograph Healthy gingiva: Low scores of BoP (under 10%); no site with PD > 3 mm; no AL
N/A
N/A
10 periodontitis patients
miRCURYTM array microarray kit, v.11.0 (Exiqon, Denmark) containing 28 to 63 yr. 1,769 miRNA capture probes 4 males 6 females
10 healthy patients
21 to 51 yr. 4 males 6 females 13 to 76 Agilent Platform (Agilent Technologies, Santa Clara, CA, USA)with the miR-451 yr. probes containing 1,205 human and 144 human viral miRNAs. miR-223 miR-486-5p miR-3917
86 periodontitis patients
3 chronic periodontitis patients
Upregulated Downregulated RT2 miRNA PCR array system (SABiosciences, Frederick, MD, USA) containing 93 inflammatory miRNA primer
let-7a let-7c miR-130a miR-301a miR-520d miR-548a
N/A
miR-126 miR-20a miR-142-3p miR-19a let-7f miR-203 miR-17 miR-223 miR-146b miR-146a
miR-155 miR-205
miR-1246 miR-1260 miR-141, miR-1260b miR-203 miR-210 miR-205
N/A
human miRNA microarray 8 15k kit (Agilent Technologies, Santa Clara, CA, USA)
miR-150 miR-223 miR-200b
miR-144 miR-379 miR-222
N/A
RT2 miRNA PCR array system (SABiosciences, Frederick, MD, USA) containing 93 inflammatory miRNA primer
miR-128 miR-34a miR-381
miR-211 miR-372 miR-656
3 edentulous residual ridges N/A
GI = gingival index; PD = probing depth; AL = attachment loss; bleeding on probing = BoP.
relate to periodontal disease, miR-146aexpression in gingival tissue shows positive correlation to other clinical criteria for chronic periodontitis including probing depth and attachment loss (Motedayyen, Ghotloo, Saffari, Sattari, & Amid, 2015). miR-207, miR-495, and miR-376b-3p are proposed as a serum biomarker in the periodontitis rat model (Tomofuji et al., 2016), but its relevance to human disease requires further investigation. The inflammation of periodontal tissue frequently results in destruction of alveolar bone and connective tissue. The microRNAs potentially have a role as a regulator in host-pathogen responses and tissue homeostasis. Many studies have reported the function of
some miRNAs in periodontitis-related inflammation. Associating with gingival epithelial cell functions, miR-105, miR-203, miR-584, and miR-128 are shown to play an important role in the modulation of innate immune response against oral pathogens. miR-105 promotes IL-6 and TNFa production by targeting toll like receptor2 (TLR2) in human primary gingival keratinocytes, when cells are stimulated with heat-inactivated P. gingivalis (Benakanakere et al., 2009). In addition, P. gingivalis induces miR-203 expression in gingival epithelial cells, which leads to STAT3 activation through the inhibition of suppressor of cytokine signaling 2 (Moffatt & Lamont, 2011). Another miRNA, miR-584, is also induced by P.
72
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
gingivalis LPS resulted in IL-8 upregulation in gingival epithelial cell line by repressing lactoferrin receptor (Ouhara et al., 2014). In human periodontal ligament cells (hPDLs), level of some miRNAs is shifted in response to P. gingivalis LPS stimulation shown by the Affimetrix array (Du et al., 2016). Subsequent validation using quantitative PCR shows that miR-21-5p, miR-498, and miR-548a-5p are significantly upregulated while miR-495-3p, miR-539-5p, and miR-7a-2-3p are downregulated. Another study in hPDLs shows that miR-146 targets IL-1 receptor-associated kinase 1 (Xie et al., 2013), thereby modulating pro-inflammatory secretion of cytokines, such as IL-1b and blocking the toll-like receptor signaling pathway after P. gingivalis LPS stimulation (Jiang et al., 2015). These results suggest a role of miRNAs in the inflammatory response to P. gingivalis infection. Recent evidence has indicated the regulatory functions of miRNAs in hPDLs involving in bone remodeling. The expression of some miRNAs is necessary for osteogenic differentiation of hPDLs. An increase of miR-132 by fluid shear stress (Qi & Zhang, 2014), or exogenous miR-146a (Hung et al., 2010), can stimulate hPDL cell proliferation and osteogenic differentiation demonstrated by alkaline phosphatase activity and in vitro mineralization. Expression of miR-17 is required in periodontal stem cells to suppress the ubiquitin regulatory factor one and promote the osteogenic differentiation of hPDLs (Liu et al., 2011). On the contrary, overexpressed miR-138 after interleukin (IL)-6 and LPS treatment reduces osteogenic differentiation in periodontal stem cells (Zhou et al., 2016), by targeting osteocalcin (OC) and Runt-related transcription factor 2 (RUNX2). The potential mRNA targets of miRNAs that have been identified in miRNA profiling studies have also been predicted. Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis have been used to define sets of genes in functional categories based on DAVID Bioinformatics Resources (Huang, Sherman, & Lempicki, 2009a; Huang, Sherman, & Lempicki, 2009b). From these analyses, 60 enriched miRNA sets have been identified, with target genes involved in immune/inflammatory responses and tissue homeostasis (Stoecklin-Wasmer et al., 2012). Interestingly, one target gene may be regulated by several miRNAs, for example, miR-218 is also shown to be able to target RUNX2 and decreases RUNX2 expression in undifferentiated human stem cells (Gay et al., 2014). Therefore, the mechanism by miRNAs appears to be tightly regulated. Although the function of miRNAs has been addressed, further studies are required to understand the miRNAs regulatory network in bone remodeling. 5. Conclusions MiRNAs regulate gene expression at the post-transcriptional level and affect numerous biological cascades. Bone metabolism, in particular, involves several miRNAs that ultimately influence cell interaction and signaling. Bone resorption may occur as a result of periodontal inflammation. RANKL expression in periodontal tissue can be induced by inflammatory mediators and leads to imbalanced bone metabolism. In periodontal resident cells, such as gingival fibroblasts and periodontal ligament cells, RANKL and inflammatory mediators can be modulated by miRNAs. In miRNA profiling studies of tissues isolated from individuals with periodontal disease, miR-223 has been consistently identified as a potential candidate miRNA to be further investigated in periodontitis-related processes. Although these studies point to an important role of miRNA-mediated epigenetic changes in tissue inflammation and alveolar bone loss, further investigation is still required to determine the function of miRNAs in the complex processes of periodontal tissue homeostasis and pathogenesis. Knowledge gained from future studies will be beneficial in
developing alternative therapeutic approaches, especially ones that use miRNA delivery systems to treat periodontal disease. References Agnihotri, R., & Gaur, S. (2015). Rheumatoid arthritis in the elderly and its relationship with periodontitis: A review. Geriatrics &. Gerontology International, 14(1), 8–22. Akhtar, N., & Haqqi, T. M. (2012). MicroRNA-199a* regulates the expression of cyclooxygenase-2 in human chondrocytes. Annals of the Rheumatic Diseases, 76 (6), 1073–1080. Almeida, M. I., Reis, R. M., & Calin, G. A. (2011). MicroRNA history: Discovery, recent applications, and next frontiers. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, 717(1), 1–8. Baek, D., Villén, J., Shin, C., Camargo, F. D., Gygi, S. P., & Bartel, D. P. (2008). The impact of microRNAs on protein output. Nature, 455(7209), 64–71. Bartel, D. P. (2009). MicroRNAs: target recognition and regulatory functions. Cell, 136 (2), 215–233. Belibasakis, G. N., Bostanci, N., Hashim, A., Johansson, A., Aduse-Opoku, J., Curtis, M. A., et al. (2007). Regulation of RANKL and OPG gene expression in human gingival fibroblasts and periodontal ligament cells by Porphyromonas gingivalis: A putative role of the Arg-gingipains. Microbial Pathogenesis, 43(1), 46–53. Benakanakere, M. R., Li, Q., Eskan, M. A., Singh, A. V., Zhao, J., Galicia, J. C., et al. (2009). Modulation of TLR2 protein expression by miR-105 in human oral keratinocytes. Journal of Biological Chemistry, 284(34), 23107–23115. Betel, D., Wilson, M., Gabow, A., Marks, D. S., & Sander, C. (2008). The microRNA. org resource: Targets and expression. Nucleic Acids Research, 36(Suppl. 1), D149– D153. Bhaumik, D., Scott, G. K., Schokrpur, S., Patil, C. K., Orjalo, A. V., Rodier, F., et al. (2009). NAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging, 1(4), 402–411. Brodersen, P., Sakvarelidze-Achard, L., Bruun-Rasmussen, M., Dunoyer, P., Yamamoto, Y. Y., Sieburth, L., et al. (2008). Widespread translational inhibition by plant miRNAs and siRNAs. Science, 320(5880), 1185–1190. Choi, B. K., Moon, S. Y., Cha, J. H., Kim, K. W., & Yoo, Y. J. (2005). Prostaglandin E2 is a main mediator in receptor activator of nuclear factor-(B ligand-dependent osteoclastogenesis induced by Porphyromonas gingivalis, Treponema denticola, and Treponema socranskii. Journal of Periodontology, 76(5), 813–820. Crockett, J. C., Rogers, M. J., Coxon, F. P., Hocking, L. J., & Helfrich, M. H. (2011). Bone remodeling at a glance. Journal of Cell Science, 124(7), 991–998. Dean, D. D., Campbell, C. M., Gruwell, S. F., Tindall, J. W., Chuang, H. H., Zhong, W., et al. (2008). Arachidonic acid and prostaglandin E2 influence human osteoblast (MG63) response to titanium surface roughness. Journal of Oral Implantology, 34 (6), 303–312. Dougall, W. C., Glaccum, M., Charrier, K., Rohrbach, K., Brasel, K., De Smedt, T., et al. (1999). RANK is essential for osteoclast and lymph node development. Genes & Development, 13(18), 2412–2424. Du, A., Zhao, S., Wan, L., Liu, T., Peng, Z., Zhou, Z., et al. (2016). MicroRNA expression profile of human periodontal ligament cells under the influence of Porphyromonas gingivalis LPS. Journal of Cellular and Molecular Medicine, 20(7), 1329–1338. Fordham, J. B., Naqvi, A. R., & Nares, S. (2015). MiR-24 regulates macrophage polarization and plasticity. Journal of Clinical & Cellular Immunology, 6(5), 362. Fraser, L. A., & Adachi, J. D. (2009). Glucocorticoid-induced osteoporosis: Treatment update and review. Therapeutic Advances in Musculoskeletal Disease, 1(2), 71–85. Gay, I., Cavender, A., Peto, D., Sun, Z., Speer, A., Cao, H., et al. (2014). Differentiation of human dental stem cells reveals a role for microRNA-218. Journal of Periodontal Research, 49(1), 110–120. Gong, M., Ma, J., Guillemette, R., Zhou, M., Yang, Y., Yang, Y., et al. (2014). MiR-335 inhibits small cell lung cancer bone metastases via IGF-IR and RANKL pathways. Molecular Cancer Research, 12(1), 101–110. Gonzalez-Martin, A., Adams, B. D., Lai, M., Shepherd, J., Salvador-Bernaldez, M., Salvador, J. M., et al. (2016). The microRNA miR-148a functions as a critical regulator of B cell tolerance and autoimmunity. Nature Immunology, 17(4), 433– 440. Grässel, S. (2014). The role of peripheral nerve fibers and their neurotransmitters in cartilage and bone physiology and pathophysiology? Arthritis Research & Therapy, 16(6), 1–13. Ha, M., & Kim, V. N. (2014). Regulation of microRNA biogenesis. Nature Reviews Molecular Cell Biology, 15(8), 509–524. Han, J., Lee, Y., Yeom, K. H., Kim, Y. K., Jin, H., & Kim, V. N. (2004). The Drosha-DGCR8 complex in primary microRNA processing. Genes & Development, 18(24), 3016– 3027. Hashizume, M., Hayakawa, N., & Mihara, M. (2008). IL-6 trans-signalling directly induces RANKL on fibroblast-like synovial cells and is involved in RANKL induction by TNF-a and IL-17. Rheumatology, 47(11), 1635–1640. Hausser, J., & Zavolan, M. (2014). Identification and consequences of miRNA-target interactions—beyond repression of gene expression. Nature Reviews Genetics, 15 (9), 599–612. Hikata, T., Takaishi, H., Takito, J., Hakozaki, A., Furukawa, M., Uchikawa, S., et al. (2009). PIAS3 negatively regulates RANKL-mediated osteoclastogenesis directly in osteoclast precursors and indirectly via osteoblasts. Blood, 113(10) [22022012].
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74 Huang, D. W., Sherman, B. T., & Lempicki, R. A. (2009a). Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Research, 37(1), 1–13. Huang, D. W., Sherman, B. T., & Lempicki, R. A. (2009b). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protocols, 4(1), 44–57. Huang, H. C., Yu, H. R., Huang, L. T., Huang, H. C., Chen, R. F., Lin, I. C., et al. (2012). MiRNA-125b regulates TNF-a production in CD14+ neonatal monocytes via post-transcriptional regulation. Journal of Leukocyte Biology, 92(1), 171–182. Hung, P. S., Chen, F. C., Kuang, S. H., Kao, S. Y., Lin, S. C., & Chang, K. W. (2010). MiR146a induces differentiation of periodontal ligament cells. Journal of Dental Research, 89(3), 252–257. Huntzinger, E., & Izaurralde, E. (2011). Gene silencing by microRNAs: Contributions of translational repression and mRNA decay. Nature Reviews Genetics, 12(2), 99– 110. Jeong, B., Kang, I., Hwang, Y., Kim, S., & Koh, J. (2014). MicroRNA-194 reciprocally stimulates osteogenesis and inhibits adipogenesis via regulating COUP-TFII expression. Cell Death & Disease, 5(11), e1532. Jiang, S. Y., Xue, D., Xie, Y. F., Zhu, D. W., Dong, Y. Y., Wei, C. C., et al. (2015). The negative feedback regulation of microRNA-146a in human periodontal ligament cells after Porphyromonas gingivalis lipopolysaccharide stimulation. Inflammation Research, 64(6), 441–451. Kalea, A., Hoteit, R., Suvan, J., Lovering, R., Palmen, J., Cooper, J., et al. (2015). Upregulation of gingival tissue miR-200b in obese periodontitis subjects. Journal of Dental Research, 94(Suppl. 3), 59S–69S. Kanzaki, H., Chiba, M., Shimizu, Y., & Mitani, H. (2002). Periodontal ligament cells under mechanical stress induce osteoclastogenesis by receptor activator of nuclear factor kB ligand up-regulation via prostaglandin E2 synthesis. Journal of Bone and Mineral Research, 17(2), 210–220. Kim, K., Lee, J., Kim, J. H., Jin, H. M., Zhou, B., Lee, S. Y., et al. (2007). Protein inhibitor of activated STAT 3 modulates osteoclastogenesis by down-regulation of NFATc1 and osteoclast-associated receptor. The Journal of Immunology, 178(9), 5588–5594. Kitaura, H., Kimura, K., Ishida, M., Kohara, H., Yoshimatsu, M., & Takano-Yamamoto, T. (2013). Immunological reaction in TNF-a-mediated osteoclast formation and bone resorption in vitro and in vivo. Clinical and Developmental Immunology2013. Kobayashi, K., Takahashi, N., Jimi, E., Udagawa, N., Takami, M., Kotake, S., et al. (2000). Tumor necrosis factor a stimulates osteoclast differentiation by a mechanism independent of the ODF/RANKL?RANK interaction. The Journal of Experimental Medicine, 191(2), 275–286. Kong, Y. Y., Yoshida, H., Sarosi, I., Tan, H. L., Timms, E., Capparelli, C., et al. (1999). OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature, 397(6717), 315–323. Krzeszinski, J. Y., Wei, W., Huynh, H., Jin, Z., Wang, X., Chang, T. C., et al. (2014). MiR34a blocks osteoporosis and bone metastasis by inhibiting osteoclastogenesis and Tgif2. Nature, 512(7515), 431–435. Kureel, J., Dixit, M., Tyagi, A., Mansoori, M., Srivastava, K., Raghuvanshi, A., et al. (2014). MiR-542-3p suppresses osteoblast cell proliferation and differentiation, targets BMP-7 signaling and inhibits bone formation. Cell Death & Disease, 5(2), e1050. Lam, J., Takeshita, S., Barker, J. E., Kanagawa, O., Ross, F. P., & Teitelbaum, S. L. (2000). TNF-a induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. The Journal of Clinical Investigation, 106(12), 1481–1488. Lee, K. N., Jang, W. G., Kim, E. J., Oh, S. H., Son, H. J., Kim, S. H., et al. (2012). Orphan nuclear receptor chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) protein negatively regulates bone morphogenetic protein 2induced osteoblast differentiation through suppressing runt-related gene 2 (Runx2) activity. Journal of Biological Chemistry, 287(23), 18888–18899. Lee, R. C., Feinbaum, R. L., & Ambros, V. (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell, 75(5), 843–854. Lee, Y. H., Na, H. S., Jeong, S. Y., Jeong, S. H., Park, H. R., & Chung, J. (2011). Comparison of inflammatory microRNA expression in healthy and periodontitis tissues. Biocell, 35(2), 43–49. Lin, D. H., Yue, P., Pan, C., Sun, P., & Wang, W. H. (2011). MicroRNA 802 stimulates ROMK channels by suppressing caveolin-1. Journal of the American Society of Nephrology, 22(6), 1087–1098. Liu, T., Qin, A. P., Liao, B., Shao, H. G., Guo, L. J., Xie, G. Q., et al. (2014). A novel microRNA regulates osteoclast differentiation via targeting protein inhibitor of activated STAT3 (PIAS3). Bone, 67, 156–165. Liu, X. H., Kirschenbaum, A., Yao, S., & Levine, A. C. (2006). Interactive Effect of Interleukin-6 and Prostaglandin E2 on Osteoclastogenesis via the OPG/RANKL/ RANK System. Annals of the New York Academy of Sciences, 1068(1), 225–233. Liu, Y., Liu, W., Hu, C., Xue, Z., Wang, G., Ding, B., et al. (2011). MiR-17 modulates osteogenic differentiation through a coherent feed-forward loop in mesenchymal stem cells isolated from periodontal ligaments of patients with periodontitis. Stem Cells, 29(11), 1804–1816. Matsuzaki, H., Wohl, G. R., Novack, D. V., Lynch, J. A., & Silva, M. J. (2007). Damaging fatigue loading stimulates increases in periosteal vascularity at sites of bone formation in the rat ulna. Calcified Tissue International, 80(6), 391–399. Matysiak, M., Fortak-Michalska, M., Szymanska, B., Orlowski, W., Jurewicz, A., & Selmaj, K. (2013). MicroRNA-146a negatively regulates the immunoregulatory activity of bone marrow stem cells by targeting prostaglandin E2 synthase-2. The Journal of Immunology, 190(10), 5102–5109.
73
McHugh, K. P., Hodivala-Dilke, K., Zheng, M. H., Namba, N., Lam, J., Novack, D., et al. (2000). Mice lacking b3 integrins are osteosclerotic because of dysfunctional osteoclasts. The Journal of Clinical Investigation, 105(4), 433–440. Melhuish, T. A., Gallo, C. M., & Wotton, D. (2001). TGIF2 interacts with histone deacetylase 1 and represses transcription. Journal of Biological Chemistry, 276 (34), 32109–32114. Miller, C. H., Smith, S. M., Elguindy, M., Zhang, T., Xiang, J. Z., Hu, X., et al. (2016). RBPJ-Regulated miR-182 promotes TNF-a?Induced osteoclastogenesis. The Journal of Immunology, 196(12), 4977–4986. Minamizaki, T., Yoshiko, Y., Kozai, K., Aubin, J. E., & Maeda, N. (2009). EP2 and EP4 receptors differentially mediate MAPK pathways underlying anabolic actions of prostaglandin E 2 on bone formation in rat calvaria cell cultures. Bone, 44(6), 1177–1185. Mine, Y., Makihira, S., Yamaguchi, Y., Tanaka, H., & Nikawa, H. (2014). Involvement of ERK and p38 MAPK pathways on Interleukin-33-induced RANKL expression in osteoblastic cells. Cell Biology International, 38(5), 655–662. Moffatt, C. E., & Lamont, R. J. (2011). Porphyromonas gingivalis induction of microRNA-203 expression controls suppressor of cytokine signaling 3 in gingival epithelial cells. Infection and Immunity, 79(7), 2632–2637. Morandini, A. C. F., Sipert, C. R., Gasparoto, T. H., Greghi, S. L. A., Passanezi, E., Rezende, M. L. R., et al. (2010). Differential production of macrophage inflammatory protein-1a, stromal-derived factor-1, and IL-6 by human cultured periodontal ligament and gingival fibroblasts challenged with lipopolysaccharide from P. gingivalis. Journal of Periodontology, 81(2), 310–317. Motedayyen, H., Ghotloo, S., Saffari, M., Sattari, M., & Amid, R. (2015). Evaluation of microRNA-146a and its targets in gingival tissues of patients with chronic periodontitis. Journal of Periodontology, 86(12), 1380–1385. Mundy, G. R. (1993). Cytokines and growth factors in the regulation of bone remodeling. Journal of Bone and Mineral Research, 8(S2), S505–S510. Murr, R. (2009). Interplay between different epigenetic modifications and mechanisms. Advances in Genetics, 70, 101–141. Na, H. S., Park, M. H., Song, Y. R., Kim, S., Kim, H. J., Lee, J. Y., et al. (2016). Elevated miR-128 in periodontitis mitigates tumor necrosis factor-alpha response via P38 signaling pathway in macrophages. Journal of Periodontology1–18. Nakasa, T., Miyaki, S., Okubo, A., Hashimoto, M., Nishida, K., Ochi, M., et al. (2008). Expression of microRNA-146 in rheumatoid arthritis synovial tissue. Arthritis & Rheumatism, 58(5), 1284–1292. Nayar, G., Gauna, A., Chukkapalli, S., Velsko, I., Kesavalu, L., & Cha, S. (2016). Polymicrobial infection alter inflammatory microRNA in rat salivary glands during periodontal disease. Anaerobe, 38, 70–75. Nebel, D., Arvidsson, J., Lillqvist, J., Holm, A., & Nilsson, B. O. (2013). Differential effects of LPS from Escherichia coli and Porphyromonas gingivalis on IL-6 production in human periodontal ligament cells. Acta Odontologica Scandinavica, 71(3-4), 892–898. Neo, W. H., Yap, K., Lee, S. H., Looi, L. S., Khandelia, P., Neo, S. X., et al. (2014). MicroRNA miR-124 controls the choice between neuronal and astrocyte differentiation by fine-tuning Ezh2 expression. Journal of Biological Chemistry, 289(30), 20788–20801. Noguchi, K., Shitashige, M., Yanai, M., Morita, I., Nishihara, T., Murota, S., et al. (1996). Prostaglandin production via induction of cyclooxygenase-2 by human gingival fibroblasts stimulated with lipopolysaccharides. Inflammation, 20(5), 555–568. Nukaga, J., Kobayashi, M., Shinki, T., Song, H., Takada, T., Takiguchi, T., et al. (2004). Regulatory effects of interleukin-1b and prostaglandin E2 on expression of receptor activator of nuclear factor-(B ligand in human periodontal ligament cells. Journal of Periodontology, 75(2), 249–259. Ogata, Y., Matsui, S., Kato, A., Zhou, L., Nakayama, Y., & Takai, H. (2014). MicroRNA expression in inflamed and noninflamed gingival tissues from Japanese patients. Journal of Oral Science, 56(4), 253–260. Ouhara, K., Savitri, I. J., Fujita, T., Kittaka, M., Kajiya, M., Iwata, T., et al. (2014). MiR584 expressed in human gingival epithelial cells is induced by Porphyromonas gingivalis stimulation and regulates interleukin-8 production via lactoferrin receptor. Journal of Periodontology, 85(6), e198–e204. Palmqvist, P., Persson, E., Conaway, H. H., & Lerner, U. H. (2002). IL-6, leukemia inhibitory factor, and oncostatin M stimulate bone resorption and regulate the expression of receptor activator of NF-kB ligand, osteoprotegerin, and receptor activator of NF-kB in mouse calvariae. The Journal of Immunology, 169(6), 3353– 3362. Parikh, A., Lee, C., Joseph, P., Marchini, S., Baccarini, A., Kolev, V., et al. (2014). microRNA -181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition. Nature Communications5. Pasquinelli, A. E. (2012). MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship. Nature Reviews Genetics, 13(4), 271–282. Perinetti, G., Paolantonio, M., D'Attilio, M., D'Archivio, D., Dolci, M., Femminella, B., et al. (2003). Aspartate aminotransferase activity in gingival crevicular fluid during orthodontic treatment. A controlled short-term longitudinal study. Journal of Periodontology, 74(2), 145–152. Perri, R., Nares, S., Zhang, S., Barros, S. P., & Offenbacher, S. (2012). MicroRNA modulation in obesity and periodontitis. Journal of Dental Research, 91(1), 33– 38. Qi, L., & Zhang, Y. (2014). The microRNA 132 regulates fluid shear stress-induced differentiation in periodontal ligament cells through mTOR signaling pathway. Cellular Physiology and Biochemistry, 33(2), 433–445. Quinn, J. M., Itoh, K., Udagawa, N., Häusler, K., Yasuda, H., Shima, N., et al. (2001). Transforming growth factor b affects osteoclast differentiation via direct and indirect actions. Journal of Bone and Mineral Research, 16(10), 1787–1794.
74
R.A. Irwandi, A. Vacharaksa / Archives of Oral Biology 72 (2016) 66–74
Raggatt, L. J., & Partridge, N. C. (2010). Cellular and molecular mechanisms of bone remodeling. Journal of Biological Chemistry, 285(33), 25103–25108. Raisz, L. G. (1999). Physiology and pathophysiology of bone remodeling. Clinical Chemistry, 45(8), 1353–1358. Rossato, M., Curtale, G., Tamassia, N., Castellucci, M., Mori, L., Gasperini, S., et al. (2012). IL-10-induced microRNA-187 negatively regulates TNF-a, IL-6, and IL12p40 production in TLR4-stimulated monocytes. Proceedings of the National Academy of Sciences, 109(45), E3101–E3110. Sánchez, G. A., Miozza, V. A., Delgado, A., & Busch, L. (2013). Salivary IL-1b and PGE2 as biomarkers of periodontal status, before and after periodontal treatment. Journal of Clinical Periodontology, 40(12), 1112–1117. Sasaki, K., Takagi, M., Konttinen, Y. T., Sasaki, A., Tamaki, Y., Ogino, T., et al. (2007). Upregulation of matrix metalloproteinase (MMP)-1 and its activator MMP-3 of human osteoblast by uniaxial cyclic stimulation. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 80(2), 491–498. Scheller, J., Chalaris, A., Schmidt-Arras, D., & Rose-John, S. (2011). The pro-and antiinflammatory properties of the cytokine interleukin-6. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, 1813(5), 878–888. Selbach, M., Schwanhäusser, B., Thierfelder, N., Fang, Z., Khanin, R., & Rajewsky, N. (2008). Widespread changes in protein synthesis induced by microRNAs. Nature, 455(7209), 58–63. Semaan, N., Frenzel, L., Alsaleh, G., Suffert, G., Gottenberg, J. E., Sibilia, J., et al. (2011). MiR-346 controls release of TNF-a protein and stability of its mRNA in rheumatoid arthritis via tristetraprolin stabilization. PLoS One, 6(5), e19827. Shah, R., Alvarez, M., Jones, D. R., Torrungruang, K., Watt, A. J., Selvamurugan, N., et al. (2004). Nmp4/CIZ regulation of matrix metalloproteinase 13 (MMP-13) response to parathyroid hormone in osteoblasts. American Journal of PhysiologyEndocrinology and Metabolism, 287(2), E289–E296. Shi, C., Qi, J., Huang, P., Jiang, M., Zhou, Q., Zhou, H., et al. (2014). MicroRNA-17/20a inhibits glucocorticoid-induced osteoclast differentiation and function through targeting RANKL expression in osteoblast cells. Bone, 68, 67–75. Singh, P., Gupta, N. D., Bey, A., & Khan, S. (2014). Salivary TNF-alpha: A potential marker of periodontal destruction. Journal of Indian Society of Periodontology, 18 (3), 306. Ståhle-Bäckdahl, M., Sandstedt, B., Bruce, K., Lindahl, A., Jiménez, M. G., Vega, J. A., et al. (1997). Collagenase-3 (MMP-13) is expressed during human fetal ossification and re-expressed in postnatal bone remodeling and in rheumatoid arthritis. Laboratory Investigation; A Journal of Technical Methods and Pathology, 76(5), 717–728. Steeve, K. T., Marc, P., Sandrine, T., Dominique, H., & Yannick, F. (2004). IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine & Growth Factor Reviews, 15(1), 49–60. Stoecklin-Wasmer, C., Guarnieri, P., Celenti, R., Demmer, R. T., Kebschull, M., & Papapanou, P. N. (2012). MicroRNAs and their target genes in gingival tissues. Journal of Dental Research, 91(10), 934–940.
Sugatani, T., & Hruska, K. A. (2009). Impaired micro-RNA pathways diminish osteoclast differentiation and function. Journal of Biological Chemistry, 284(7), 4667–4678. Taganov, K. D., Boldin, M. P., Chang, K. J., & Baltimore, D. (2006). NF-(B-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences, 103 (33), 12481–12486. Tomofuji, T., Yoneda, T., Machida, T., Ekuni, D., Azuma, T., Kataoka, K., et al. (2016). MicroRNAs as serum biomarkers for periodontitis. Journal of Clinical Periodontology, 43(5), 418–425. Udagawa, N., Takahashi, N., Jimi, E., Matsuzaki, K., Tsurukai, T., Itoh, K., et al. (1999). Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colony-stimulating factor. Bone, 25(5), 517–523. Varki, A., & Altheide, T. K. (2005). Comparing the human and chimpanzee genomes: Searching for needles in a haystack. Genome Research, 15(12), 1746–1758. Wada, T., Nakashima, T., Hiroshi, N., & Penninger, J. M. (2006). RANKL–RANK signaling in osteoclastogenesis and bone disease. Trends in Molecular Medicine, 12(1), 17–25. Wang, T., Yin, H., Wang, J., Li, Z., Wei, H., Liu, Z. A., et al. (2015). MicroRNA-106b inhibits osteoclastogenesis and osteolysis by targeting RANKL in giant cell tumor of bone. Oncotarget, 6(22), 18980–18996. Wang, X. (2014). Composition of seed sequence is a major determinant of microRNA targeting patterns. Bioinformatics, 30(10), 1377–1383. Xie, Y. F., Shu, R., Jiang, S. Y., Liu, D. L., & Zhang, X. L. (2011). Comparison of microRNA profiles of human periodontal diseased and healthy gingival tissues. International Journal of Oral Science, 3(3), 125–134. Xie, Y. F., Shu, R., Jiang, S. Y., Liu, D. L., Ni, J., & Zhang, X. L. (2013). MicroRNA-146 inhibits pro-inflammatory cytokine secretion through IL-1 receptor-associated kinase 1 in human gingival fibroblasts. J Inflamm (Lond), 10(1), 20–28. Xie, Z., Yin, X., Gong, B., Nie, W., Wu, B., Zhang, X., et al. (2015). Salivary microRNAs show potential as a noninvasive biomarker for detecting resectable pancreatic cancer. Cancer Prevention Research, 8(2), 165–173. Yang, C. M., Chien, C. S., Yao, C. C., Hsiao, L. D., Huang, Y. C., & Wu, C. B. (2004). Mechanical strain induces collagenase-3 (MMP-13) expression in MC3T3-E1 osteoblastic cells. Journal of Biological Chemistry, 279(21), 22158–22165. Zhang, Y. H., Heulsmann, A., Tondravi, M. M., Mukherjee, A., & Abu-Amer, Y. (2001). Tumor necrosis factor-a (TNF) stimulates RANKL-induced osteoclastogenesis via coupling of TNF type 1 receptor and RANK signaling pathways. Journal of Biological Chemistry, 276(1), 563–568. Zhou, X., Luan, X., Chen, Z., Francis, M., Gopinathan, G., Li, W., et al. (2016). MicroRNA-138 inhibits periodontal progenitor differentiation under inflammatory conditions. Journal of Dental Research, 95(2), 230–237. Zou, W., Hakim, I., Tschoep, K., Endres, S., & Bar-Shavit, Z. (2001). Tumor necrosis factor-a mediates RANK ligand stimulation of osteoclast differentiation by an autocrine mechanism. Journal of Cellular Biochemistry, 83(1), 70–83.