Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2067 word(s) 2067 2022-01-24 02:34:06

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Bhawal, U. Fluoride (NaF) in Dentistry. Encyclopedia. Available online: https://encyclopedia.pub/entry/18805 (accessed on 29 July 2024).
Bhawal U. Fluoride (NaF) in Dentistry. Encyclopedia. Available at: https://encyclopedia.pub/entry/18805. Accessed July 29, 2024.
Bhawal, Ujjal. "Fluoride (NaF) in Dentistry" Encyclopedia, https://encyclopedia.pub/entry/18805 (accessed July 29, 2024).
Bhawal, U. (2022, January 26). Fluoride (NaF) in Dentistry. In Encyclopedia. https://encyclopedia.pub/entry/18805
Bhawal, Ujjal. "Fluoride (NaF) in Dentistry." Encyclopedia. Web. 26 January, 2022.
Fluoride (NaF) in Dentistry
Edit

Fluoride is well known for its use in the treatment of dental caries, either systemically or topically. Fluoride intake (such as in drinking water, fluoridated toothpaste, or fluoride supplements) is the cornerstone to preventing dental caries in adults and children. Fluoride prevents dental caries by slowing down the demineralization of enamel, which is caused by the interaction between dental plaque and dental hard tissues. Fluoride may inhibit tooth decay by 40–60% by co-precipitating calcium and phosphate ions and by enhancing the precipitation of fluoridated apatite. Fluoride is also found deposited as calcium fluoride in dental plaque, which helps to prevent dental caries.

fluoride dentistry osteoblasts osteoclasts

1. Fluoride and Epithelial Cells

Epithelial cells function as a barrier to protect organisms from physical, chemical, and microbial damage and to maintain mucosal integrity. Dental epithelial cells can differentiate into various types of cells that can secrete enamel matrix proteins during tooth development [1]. The matrix metalloproteinase-20 (MMP-20) is produced by ameloblasts and is responsible for cleaving other enamel matrix proteins such as amelogenin, ameloblastin, and enamelin [2]. MMP-20 is produced at the secretory stage of dental enamel formation and cleaves proteins that allow the crystals to grow into the space [2]. Ameloblasts also secrete kallikrein-4 (KLK4), which is essential for the breakdown of enamel proteins and enamel crystallization [3]. KLK4 helps crystals to grow and contract, which strengthens the enamel at the last stage of amelogenesis [3].
Recent studies have revealed that different concentrations of fluoride have various effects on epithelial cells.

1.1. High Concentration Fluoride and Epithelial Cells

A high concentration of NaF (millimolar level) induces endoplasmic reticulum stress, apoptosis, and DNA fragmentation and interferes with enamel proteinases [4]. Exposure to a high concentration of fluoride results in enamel hypomineralization, also known as enamel fluorosis [5]. The severity of enamel fluorosis increases depending on the volume of fluoride intake and the duration of fluoride exposure [5]. Fluoride decreases the abilities of proteinases such as MMP-20 and KLK4 that degrade enamel matrix proteins, and as a result, inhibit crystal growth [6]. Alternatively, the intake of fluoride lowers the pH, which inhibits the synthesis and secretion of KLK4 [7]. One study reported that excessive fluoride results in hypomineralization through the reduced expression of Forkhead box O1 (FOXO1) in dental epithelial cells [8]. FOXO1 is reduced after exposure to excessive fluoride, which might affect the expressions of KLK4 and amelotin (AMTN), which ultimately results in enamel fluorosis. One mM fluoride was found to be cytotoxic and induced apoptosis in human gingival epithelial cells. Treatment with 2 mM NaF disturbed the gene network accompanied by endoplasmic reticulum stress [9].

1.2. Low Concentration Fluoride and Epithelial Cells

Studies have shown that fluoride has biphasic effects on epithelial ameloblast-lineage cells at different concentrations. Reduced proliferation was observed at a fluoride concentration higher than 1 mM; however, lower concentrations of fluoride, e.g., around 16 μM, promoted the proliferation of epithelial ameloblast-lineage cells [10].
During gingival wound healing, human epithelial cells cover the wound, a process termed re-epithelialization. The process of wound healing includes epithelial cell proliferation and migration as well as the synthesis and deposition of extracellular matrix components [11]. Extracellular matrix proteins such as fibronectin and laminin-5 are expressed by migrating keratinocytes during wound healing. To understand the characteristics of gingival epithelial cells responding to fluoride, human primary epithelial cells were treated with NaF to characterize their effects on cellular physiology. Cell proliferation peaked at a concentration of 50 μM of NaF, and a higher concentration of NaF (at the millimolar level) reduced proliferation. Cells treated with 50 μM of NaF showed significantly more motility than non-treated cells in vitro. qRT-PCR analyses showed increased mRNA expression levels of fibronectin and laminin-5 in cells treated with 50 μM of NaF [12].

1.3. Fluoride and Epithelial-Mesenchymal Interactions

Epithelial-mesenchymal interactions (EMT) are important for the development of ectodermal organs and for tissue regeneration and wound healing, including cellular events such as cell adhesion, proliferation, differentiation, and maturation [13]. Homeostasis, inflammation, migration and proliferation, and remodeling are the four major processes of wound healing [14]. Fibroblast growth factor 2 (FGF2) and its receptor, fibroblast growth factor receptor 2 (FGFR2), are crucial for proliferation, migration, and protease production in epithelial cells [15]. FGF2 is associated with EMT by inducing mesenchymal characteristics in epithelial cells [16]. Fibroblast growth factor 7 (FGF7) is essential for epithelial morphogenesis [17]. Twist family BHLH transcription factor 1 (Twist1) is a critical mediator for EMT [18]. One study demonstrated that treatment with 50 μM of NaF induces the expression of FGF2 and FGF7, and Twist1 is also upregulated in vivo. It was also found that the untreated group had more persistent wounds in mice [19].

2. Fluoride and Bone Marrow Mesenchymal Stem Cells (BMMSCs)

BMMSCs present in the bone marrow is self-renewing precursor cells that have the multipotency to differentiate into osteoblasts, chondroblasts, adipoblasts, and stromal cells [20]. Upon bone injuries, BMMSCs differentiate into osteoblasts and release growth factors during wound healing [21]. Consistent with other studies, fluoride also has dual effects on BMMSCs. A low concentration of NaF (50 or 500 μM) enhanced the proliferation of BMMSCs, while a high concentration of NaF (5 mM) reduced their proliferation. Furthermore, BMMSCs showed elevated motility and migration when compared with the control group. Treatment with 50 μM of NaF upregulated the expression of fibronectin and vimentin, which induced the Runt-related transcription factor (Runx2), induced osteoblast differentiation, and increased the secretion of osteocalcin (OCN) [22]. Treatment with a higher concentration of NaF induced cytotoxicity, DNA damage, and oxidative stress in BMMSCs [23]. Studies have indicated that fluoride-induced cytotoxicity depends on the concentration and the duration of fluoride exposure [24]. High concentrations of NaF (2 mM and above) result in oxidative stress and decreased viability and proliferation via the phosphor-c-Jun N-terminal kinase (JNK) pathway. In dental pulp stem cells, a low concentration of NaF promotes osteo/odontogenic differentiation via the PI3K/Akt pathway [25]. In addition, a low concentration of NAF (0.5 mM) is the optimal concentration to regulate the osteo/odontogenic differentiation of stem cells from apical papilla [26]. When exposed to mouse embryonic stem cells, a high concentration of NaF (over 1 mM) induced ROS and reduced DNA synthesis, which resulted in apoptosis through a JNK-dependent pathway [27].

3. Fluoride and Bone Metabolism

Bone homeostasis is maintained through the bone formation by osteoblasts and bone resorption by osteoclasts [28]. The activity of osteoblasts and osteoclasts is critical for bone maintenance and remodeling. The process is regulated by many factors, such as sex hormones, parathyroid hormones, and calcitonin, as well as various growth factors and cytokines [29]. Osteoblasts and osteoclasts also can control each other’s formation, differentiation, apoptosis through multiple pathways via cytokines, extracellular proteins, and transcription factors [30]. Bone diseases such as osteoporosis are caused by dysfunctional bone remodeling or the disruption of the homeostasis maintained by osteoblasts and osteoclasts [30].
Both positive and negative effects of fluoride on bones and teeth have been well established [31][32]. Fluoride treatment increases the proliferation of osteoblasts and inhibits the function of osteoclasts [33]. Trace amounts of fluoride have been used to treat osteoporosis, vertebral fractures, and bone loss in patients [34], whereas excessive fluoride results in skeletal fluorosis [35]. Long-term exposure to fluoride can result in skeletal and dental fluorosis [36]. Thus, a better understanding of the effects of different concentrations of NaF on bone homeostasis will help to gain more insights into the usage of fluoride.

3.1. Fluoride and Osteoblasts

OCN and Osteopontin (OPN) are non-collagenous proteins secreted by osteoblasts that serve as markers for osteoblast maturation [37]. OCN promotes bone formation and regulates mineralization in the bone matrix, which has a complex regulatory network [38]. OPN is an extracellular matrix protein that has multiple functions associated with bone structuring and destruction in osseous tissues [39]. The transcription factor Runx2 induces the expression of osteoblastic genes including OCN and OPN [40]. Runx2 is expressed at different stages of osteoblasts (pre-osteoblasts, and immature and mature osteoblasts) and is essential for bone formation and osteoblastic differentiation [41]. Osterix (OSX), a zinc-finger containing a transcription factor, is also a downstream target of Runx2 [42]. Together with Runx2, OSX regulates the differentiation of pre-osteoblasts into mature osteoblasts and osteocytes, and the roles of OSX in maintaining bone homeostasis have been well studied [24]. One study showed that NaF treatment affects calcium homeostasis and transcription factor expression [43]. A low concentration of NaF induced Runx2 and OSX while a high concentration of NaF reduced their expression both at the mRNA and protein levels. Treatment of MC3T3-E1 cells with 50 or 500 μM of NaF enhanced their proliferation, alkaline phosphatase (ALP) activity, and extracellular matrix mineralization, as well as their expression of bone-related genes (Runx2, OSX, OCN, and OPN) [44]. Different concentrations of NaF were also found to differentially affect the expression of bone mineralized regulator proteins, such as OCN, OPN, osteonectin (ON), and bone sialoprotein (BSP) in bone marrow stromal cells. A higher concentration of NaF (10−5 M) decreased the level of the proteins while a lower concentration (10−7 M) increased them [31].
Various cellular mechanisms, including the Mitogen-activated protein kinase (MAPK) pathway, are proposed to function in bone formation during fluoride treatment [45]. It is well established that a low concentration of NaF (micromolar levels) regulates tyrosine kinase and ALP activity to increase osteoblast proliferation [46][47]. Fluoride treatment of osteoblasts is also biphasic in that low concentrations promote proliferation, whereas higher concentrations result in signs of weakened osteoblast activity [48]. Long periods of fluoride exposure reduce the expression of bcl-2 family proteins, which promotes apoptosis in osteoblastic cells [49]. Fluoride induces mitochondrial respiratory chain complex abnormal expressions, which in turn cause oxidative stress and result in apoptosis [50]. High concentrations of NaF also regulate the proliferation and cell cycle of the p16 gene methylation during the development of skeletal fluorosis [51]. The schematic diagram shown in Figure 1 summarizes the functions of NaF in osteoblasts.
Figure 1. A schematic diagram of the biphasic functions of fluoride in osteoblasts. A low concentration of NaF stimulates the expression of fibronectin, vimentin, Runx2, and OSX to promote the expression of osteoblastic genes (OCN and OPN). A high concentration of NaF inhibits Runx2 and induces oxidative stress in osteoblasts.

3.2. Fluoride and Osteoclasts

Osteoprotegerin (OPG) is a soluble cytokine receptor of the tumor necrosis factor (TNF) receptor family that binds to the OPG ligand [52]. The receptor activator of nuclear factor kappa-B ligand (RANKL) is also a member of the TNF superfamily, which binds to RANK on target cells. OPG/RANKL/RANK is a key signaling pathway in bone metabolism. The overexpression of OPG alters osteoclast differentiation, and recombinant OPG impedes ovariectomy-induced bone loss in rats [53]. Recombinant OPG binds to OPG ligand on BMMSCs, thereby inhibiting osteoclast differentiation [54]. RANKL is an essential cytokine that regulates osteoclastogenesis and bone resorption [55]. In response to RANKL activation, the nuclear factor of activated T cells 1 (NFATc1) regulates the terminal differentiation of osteoclasts, and NFATc1-deficient embryonic stem cells were unable to differentiate into osteoclasts [56]. NFATc1 participates in the transcription of the genes involved in heart/valve septum formation, angiogenesis, T cell proliferation, and osteoclast formation. NFATc1 binds to the promoter of genes associated with bone resorption, including cathepsin K and matrix metalloproteinase 9 (MMP-9), thus inducing their gene expression [57].
Low concentrations of NaF (micromolar level) have little influence on the viability of BMMSCs and significantly downregulate both mRNA and the protein expression levels of NFATc1 in rat osteoclasts, which result in a reduction in the levels of cathepsin K and the attenuation of bone destruction [58]. Treatment with 0.5 mM to 1 mM of NaF inhibits the activity of osteoclasts in vitro [59]. Studies suggest that fluoride can act on matrix proteinases such as Metalloproteinases-2 and -9, thus inhibiting matrix degradation [60][61]. Low concentrations of fluoride can possibly regulate osteoclasts via the B lymphocyte-induced maturation protein-1 (Blimp1)/B cell lymphoma 6 (Bcl6) axis which is a critical signaling pathway that regulates osteoclast differentiation and bone homeostasis [62]. On the other hand, the stimulation of NaF exhibits a U-shaped curve in a dose-dependent manner [63]. High concentrations of NaF induce RANKL and decrease OPG, thus increasing osteocyte-driven osteoclastogenesis via the RANK-JNK-NFATc1 signaling pathway [64]. A recent study found that osteoclasts demonstrate the most sensitivity to high concentrations of NaF with respect to other bone cell types and that fluoride exposure induces apoptosis via the Transforming growth factor (TGF)-β signaling pathway [65]. A schematic diagram of the functions of NaF in osteoclasts is shown in Figure 2.
Figure 2. A schematic diagram of the biphasic functions of fluoride in osteoclasts. A low concentration of NaF inhibits the expression of RANKL, NFATc1, MMP9, and Cathepsin K but induces OPG to bind RANKL. A low concentration of NaF is possibly associated with Blimp1/Bcl6 to repress the expression of osteoclastic genes (MMP9 and Cathepsin K). A high concentration of NaF decreases the expression of OPG while inducing RANKL to bind RANK. Induced NFATc1 translocate into the nucleus to promote osteoclastogenesis.

References

  1. Kawano, S.; Saito, M.; Handa, K.; Morotomi, T.; Toyono, T.; Seta, Y.; Nakamura, N.; Uchida, T.; Toyoshima, K.; Ohishi, M.; et al. Characterization of Dental Epithelial Progenitor Cells Derived from Cervical-loop Epithelium in a Rat Lower Incisor. J. Dent. Res. 2004, 83, 129–133.
  2. Crivelini, M.M.; Oliveira, D.T.; De Mesquita, R.A.; De Sousa, S.C.O.M.; Loyola, A.M. Kallikrein 4 and matrix metalloproteinase-20 immunoexpression in malignant, benign and infiltrative odontogenic tumors. J. Oral Maxillofac. Pathol. 2016, 20, 246–251.
  3. Mu, Y.; Tian, R.; Xiao, L.; Sun, D.; Zhang, Z.; Xu, S.; Yang, G. Molecular Evolution of Tooth-Related Genes Provides New Insights into Dietary Adaptations of Mammals. J. Mol. Evol. 2021, 89, 458–471.
  4. Wei, W.; Gao, Y.; Wang, C.; Zhao, L.; Sun, D. Excessive fluoride induces endoplasmic reticulum stress and interferes enamel proteinases secretion. Environ. Toxicol. 2011, 28, 332–341.
  5. DenBesten, P.K.; Zhu, L.; Li, W.; Tanimoto, K.; Liu, H.; Witkowska, H.E. Fluoride incorporation into apatite crystals delays amelogenin hydrolysis. Eur. J. Oral Sci. 2011, 119, 3–7.
  6. DenBesten, P.K.; Yan, Y.; Featherstone, J.D.B.; Hilton, J.F.; Smith, C.E.; Li, W. Effects of fluoride on rat dental enamel matrix proteinases. Arch. Oral Biol. 2002, 47, 763–770.
  7. Sharma, R.; Tsuchiya, M.; Skobe, Z.; Tannous, B.A.; Bartlett, J.D. The Acid Test of Fluoride: How pH Modulates Toxicity. PLoS ONE 2010, 5, e10895.
  8. Gao, J.; Ruan, J.; Gao, L. Excessive fluoride reducesFoxo1expression in dental epithelial cells of the rat incisor. Eur. J. Oral Sci. 2014, 122, 317–323.
  9. Zuckerbraun, H.L.; Babich, H.; May, R.; Sinensky, M.C. Triclosan, cytotoxicity, mode of action, and induction of apoptosis in human gingival cells in vitro. Eur. J. Oral Sci. 1998, 106, 628–636.
  10. Yan, Q.; Zhang, Y.; Li, W.; DenBesten, P. Micromolar Fluoride Alters Ameloblast Lineage Cells in vitro. J. Dent. Res. 2007, 86, 336–340.
  11. Jorgensen, S.N.; Sanders, J.R. Mathematical models of wound healing and closure: A comprehensive review. Med. Biol. Eng. Comput. 2015, 54, 1297–1316.
  12. Arakawa, Y.; Bhawal, U.K.; Ikoma, T.; Kimoto, K.; Kuroha, K.; Kubota, T.; Hamada, N.; Kubota, E.; Arakawa, H. Low concentration fluoride stimulates cell motility of epithelial cells in vitro. Biomed. Res. 2009, 30, 271–277.
  13. Wixler, V. The role of FHL2 in wound healing and inflammation. FASEB J. 2019, 33, 7799–7809.
  14. Cho, Y.-D.; Kim, K.-H.; Lee, Y.-M.; Ku, Y.; Seol, Y.-J. Periodontal Wound Healing and Tissue Regeneration: A Narrative Review. Pharmaceuticals 2021, 14, 456.
  15. Mossahebi-Mohammadi, M.; Quan, M.; Zhang, J.-S.; Li, X. FGF Signaling Pathway: A Key Regulator of Stem Cell Pluripotency. Front. Cell Dev. Biol. 2020, 8, 79.
  16. El-Baz, L.M.; Shoukry, N.M.; Hafez, H.S.; Guzy, R.D.; Salem, M.L. Fibroblast Growth Factor 2 Augments Transforming Growth Factor Beta 1 Induced Epithelial-mesenchymal Transition in Lung Cell Culture Model. Iran. J. Allergy Asthma Immunol. 2020, 19, 348–361.
  17. Kinoshita-Ise, M.; Tsukashima, A.; Kinoshita, T.; Yamazaki, Y.; Ohyama, M. Altered FGF expression profile in human scalp-derived fibroblasts upon WNT activation: Implication of their role to provide folliculogenetic microenvironment. Inflamm. Regen. 2020, 40, 1–10.
  18. Meng, J.; Chen, S.; Han, J.-X.; Qian, B.; Wang, X.-R.; Zhong, W.-L.; Qin, Y.; Zhang, H.; Gao, W.-F.; Lei, Y.-Y.; et al. Twist1 Regulates Vimentin through Cul2 Circular RNA to Promote EMT in Hepatocellular Carcinoma. Cancer Res. 2018, 78, 4150–4162.
  19. He, D.; Bhawal, U.K.; Hamada, N.; Kuboyama, N.; Abiko, Y.; Arakawa, H. Low Level Fluoride Stimulates Epithelial-Mesenchymal Interaction in Oral Mucosa. J. Hard Tissue Biol. 2013, 22, 59–66.
  20. Chen, F.; Han, Y.; Kang, Y. Bone marrow niches in the regulation of bone metastasis. Br. J. Cancer 2021, 214, 1912–1920.
  21. Zhang, R.; Li, X.; Liu, Y.; Gao, X.; Zhu, T.; Lu, L. Acceleration of Bone Regeneration in Critical-Size Defect Using BMP-9-Loaded nHA/ColI/MWCNTs Scaffolds Seeded with Bone Marrow Mesenchymal Stem Cells. BioMed Res. Int. 2019, 2019, 7343957.
  22. Bhawal, U.K.; Li, X.; Suzuki, M.; Taguchi, C.; Oka, S.; Arikawa, K.; Tewari, N.; Liu, Y. Treatment with low-level sodium fluoride on wound healing and the osteogenic differentiation of bone marrow mesenchymal stem cells. Dent. Traumatol. 2019, 36, 278–284.
  23. Garcia, A.L.; Picinini, J.; Silveira, M.D.; Camassola, M.; Visentim, A.P.; Salvador, M.; da Silva, J. Fluorosilicic acid induces DNA damage and oxidative stress in bone marrow mesenchymal stem cells. Mutat. Res. Toxicol. Environ. Mutagen. 2020, 861–862, 503297.
  24. Fu, X.; Xie, F.-N.; Dong, P.; Li, Q.-C.; Yu, G.-Y.; Xiao, R. High-Dose Fluoride Impairs the Properties of Human Embryonic Stem Cells via JNK Signaling. PLoS ONE 2016, 11, e0148819.
  25. Xu, S.; Xie, X.; Li, C.; Liu, Z.; Zuo, D. Micromolar sodium fluoride promotes osteo/odontogenic differentiation in dental pulp stem cells by inhibiting PI3K/AKT pathway. Arch. Oral Biol. 2021, 131, 105265.
  26. Pan, Y.; Li, Z.; Wang, Y.; Yan, M.; Wu, J.; Beharee, R.G.; Yu, J. Sodium fluoride regulates the osteo/odontogenic differentiation of stem cells from apical papilla by modulating autophagy. J. Cell. Physiol. 2019, 234, 16114–16124.
  27. Ngoc, T.D.N.; Son, Y.-O.; Lim, S.-S.; Shi, X.; Kim, J.-G.; Heo, J.S.; Choe, Y.; Jeon, Y.-M.; Lee, J.-C. Sodium fluoride induces apoptosis in mouse embryonic stem cells through ROS-dependent and caspase- and JNK-mediated pathways. Toxicol. Appl. Pharmacol. 2012, 259, 329–337.
  28. Siddiqui, J.A.; Partridge, N.C. Physiological Bone Remodeling: Systemic Regulation and Growth Factor Involvement. Physiology 2016, 31, 233–245.
  29. Chen, X.; Wang, Z.; Duan, N.; Zhu, G.; Schwarz, E.M.; Xie, C. Osteoblast–osteoclast interactions. Connect. Tissue Res. 2017, 59, 99–107.
  30. Sims, N.A.; Martin, T.J. Osteoclasts Provide Coupling Signals to Osteoblast Lineage Cells Through Multiple Mechanisms. Annu. Rev. Physiol. 2020, 82, 507–529.
  31. Antonarakis, G.S.; Moseley, R.; Waddington, R.J. Differential influence of fluoride concentration on the synthesis of bone matrix glycoproteins within mineralizing bone cellsin vitro. Acta Odontol. Scand. 2014, 72, 1066–1069.
  32. Simon, M.J.K.; Beil, F.T.; Riedel, C.; Lau, G.; Tomsia, A.; Zimmermann, E.A.; Koehne, T.; Ueblacker, P.; Rüther, W.; Pogoda, P.; et al. Deterioration of teeth and alveolar bone loss due to chronic environmental high-level fluoride and low calcium exposure. Clin. Oral Investig. 2016, 20, 2361–2370.
  33. Liu, S.; Zhou, H.; Liu, H.; Ji, H.; Fei, W.; Luo, E. Fluorine-contained hydroxyapatite suppresses bone resorption through inhibiting osteoclasts differentiation and function in vitro and in vivo. Cell Prolif. 2019, 52, e12613.
  34. Yamaguchi, M. Fluoride and bone metabolism. Clin. Calcium 2007, 17, 217–223.
  35. Kurdi, M.S. Chronic fluorosis: The disease and its anaesthetic implications. Indian J. Anaesth. 2016, 60, 157–162.
  36. Qiao, W.; Liu, Q.; Li, Z.; Zhang, H.; Chen, Z. Changes in physicochemical and biological properties of porcine bone derived hydroxyapatite induced by the incorporation of fluoride. Sci. Technol. Adv. Mater. 2017, 18, 110–121.
  37. Epsley, S.; Tadros, S.; Farid, A.; Kargilis, D.; Mehta, S.; Rajapakse, C.S. The Effect of Inflammation on Bone. Front. Physiol. 2021, 11.
  38. Zoch, M.L.; Clemens, T.L.; Riddle, R.C. New insights into the biology of osteocalcin. Bone 2015, 82, 42–49.
  39. Icer, M.A.; Gezmen-Karadag, M. The multiple functions and mechanisms of osteopontin. Clin. Biochem. 2018, 59, 17–24.
  40. Tang, C.-Y.; Wu, M.; Zhao, D.; Edwards, D.; McVicar, A.; Luo, Y.; Zhu, G.; Wang, Y.; Zhou, H.-D.; Chen, W.; et al. Runx1 is a central regulator of osteogenesis for bone homeostasis by orchestrating BMP and WNT signaling pathways. PLoS Genet. 2021, 17, e1009233.
  41. Takahata, Y.; Hagino, H.; Kimura, A.; Urushizaki, M.; Kobayashi, S.; Wakamori, K.; Fujiwara, C.; Nakamura, E.; Yu, K.; Kiyonari, H.; et al. Smoc1 and Smoc2 regulate bone formation as downstream molecules of Runx2. Commun. Biol. 2021, 4, 1–11.
  42. Iaquinta, M.R.; Lanzillotti, C.; Mazziotta, C.; Bononi, I.; Frontini, F.; Mazzoni, E.; Oton-Gonzalez, L.; Rotondo, J.C.; Torreggiani, E.; Tognon, M.; et al. The role of microRNAs in the osteogenic and chondrogenic differentiation of mesenchymal stem cells and bone pathologies. Theranostics 2021, 11, 6573–6591.
  43. Duan, X.-Q.; Zhao, Z.-T.; Zhang, X.-Y.; Wang, Y.; Wang, H.; Liu, D.-W.; Li, G.-S.; Jing, L. Fluoride Affects Calcium Homeostasis and Osteogenic Transcription Factor Expressions Through L-type Calcium Channels in Osteoblast Cell Line. Biol. Trace Element Res. 2014, 162, 219–226.
  44. Lee, M.; Arikawa, K.; Nagahama, F. Micromolar Levels of Sodium Fluoride Promote Osteoblast Differentiation Through Runx2 Signaling. Biol. Trace Element Res. 2017, 178, 283–291.
  45. Volobaev, V.P.; Serdyukova, E.S.; Kalyuzhnaya, E.E.; Schetnikova, E.A.; Korotkova, A.D.; Naik, A.A.; Bach, S.N.; Prosekov, A.Y.; Larionov, A.V. Investigation of the genotoxic effects of fluoride on a bone tissue model. Toxicol. Res. 2020, 36, 337–342.
  46. Burgener, D.; Bonjour, J.-P.; Caverzasio, J. Fluoride increases tyrosine kinase activity in osteoblast-like cells: Regulatory role for the stimulation of cell proliferation and Pi transport across the plasma membrane. J. Bone Miner. Res. 2009, 10, 164–171.
  47. Farley, J.R.; Wergedal, J.E.; Baylink, D.J. Fluoride Directly Stimulates Proliferation and Alkaline Phosphatase Activity of Bone-Forming Cells. Science 1983, 222, 330–332.
  48. Pak, C.Y.; Zerwekh, J.E.; Antich, P. Anabolic effects of fluoride on bone. Trends Endocrinol. Metab. 1995, 6, 229–234.
  49. Rochette, L.; Meloux, A.; Rigal, E.; Zeller, M.; Cottin, Y.; Vergely, C. The Role of Osteoprotegerin and Its Ligands in Vascular Function. Int. J. Mol. Sci. 2019, 20, 705.
  50. Yang, S.; Wang, Z.; Farquharson, C.; Alkasir, R.; Zahra, M.; Ren, G.; Han, B. Sodium fluoride induces apoptosis and alters bcl-2 family protein expression in MC3T3-E1 osteoblastic cells. Biochem. Biophys. Res. Commun. 2011, 410, 910–915.
  51. Zhao, W.-P.; Wang, H.-W.; Liu, J.; Zhang, Z.-H.; Zhu, S.-Q.; Zhou, B.-H. Mitochondrial respiratory chain complex abnormal expressions and fusion disorder are involved in fluoride-induced mitochondrial dysfunction in ovarian granulosa cells. Chemosphere 2018, 215, 619–625.
  52. Wu, S.; Yan, W.; Qiu, B.; Liao, Y.; Gu, J.; Wei, S.; Zhang, A.; Pan, X. Aberrant methylation-induced dysfunction of p16 is associated with osteoblast activation caused by fluoride. Environ. Toxicol. 2018, 34, 37–47.
  53. Simonet, W.; Lacey, D.; Dunstan, C.; Kelley, M.; Chang, M.-S.; Lüthy, R.; Nguyen, H.; Wooden, S.; Bennett, L.; Boone, T.; et al. Osteoprotegerin: A Novel Secreted Protein Involved in the Regulation of Bone Density. Cell 1997, 89, 309–319.
  54. Usui, M.; Onizuka, S.; Sato, T.; Kokabu, S.; Ariyoshi, W.; Nakashima, K. Mechanism of alveolar bone destruction in periodontitis—Periodontal bacteria and inflammation. Jpn. Dent. Sci. Rev. 2021, 57, 201–208.
  55. McDonald, M.M.; Kim, A.S.; Mulholland, B.S.; Rauner, M. New Insights Into Osteoclast Biology. JBMR Plus 2021, 5, e10539.
  56. Zhao, X.; Patil, S.; Xu, F.; Lin, X.; Qian, A. Role of Biomolecules in Osteoclasts and Their Therapeutic Potential for Osteoporosis. Biomolecules 2021, 11, 747.
  57. Kitamura, N.; Kaminuma, O. Isoform-Selective NFAT Inhibitor: Potential Usefulness and Development. Int. J. Mol. Sci. 2021, 22, 2725.
  58. Bhawal, U.K.; Lee, H.-J.; Arikawa, K.; Shimosaka, M.; Suzuki, M.; Toyama, T.; Sato, T.; Kawamata, R.; Taguchi, C.; Hamada, N.; et al. Micromolar sodium fluoride mediates anti-osteoclastogenesis in Porphyromonas gingivalis-induced alveolar bone loss. Int. J. Oral Sci. 2015, 7, 242–249.
  59. Okuda, A.; Kanehisa, J.; Heersche, J.N.M. The effects of sodium fluoride on the resorptive activity of isolated osteoclasts. J. Bone Miner. Res. 2010, 5, S115–S120.
  60. Łukomska, A.; Baranowska-Bosiacka, I.; Dec, K.; Pilutin, A.; Tarnowski, M.; Jakubczyk, K.; Żwierełło, W.; Skórka-Majewicz, M.; Chlubek, D.; Gutowska, I. Changes in Gene and Protein Expression of Metalloproteinase-2 and -9 and their Inhibitors TIMP2 and TIMP3 in Different Parts of Fluoride-Exposed Rat Brain. Int. J. Mol. Sci. 2020, 22, 391.
  61. Cvikl, B.; Lussi, A.; Carvalho, T.S.; Moritz, A.; Gruber, R. Stannous chloride and stannous fluoride are inhibitors of matrix metalloproteinases. J. Dent. 2018, 78, 51–58.
  62. 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.
  63. Yao, Y.; Ma, Y.; Zhong, N.; Pei, J. The Inverted U-Curve Association of Fluoride and Osteoclast Formation in Mice. Biol. Trace Elem. Res. 2019, 191, 419–425.
  64. Jiang, N.; Guo, F.; Xu, W.; Zhang, Z.; Jin, H.; Shi, L.; Zhang, X.; Gao, J.; Xu, H. Effect of fluoride on osteocyte-driven osteoclastic differentiation. Toxicology 2020, 436, 152429.
  65. Jiang, N.; Guo, F.; Sun, B.; Zhang, X.; Xu, H. Different Effects of Fluoride Exposure on the Three Major Bone Cell Types. Biol. Trace Elem. Res. 2019, 193, 226–233.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 502
Revision: 1 time (View History)
Update Date: 26 Jan 2022
1000/1000
Video Production Service