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 -- 1890 2022-05-09 08:20:54 |
2 format correct -3 word(s) 1887 2022-05-09 08:28:18 | |
3 Some contents have been added. + 407 word(s) 2294 2022-05-09 08:35:07 | |
4 format correct + 11 word(s) 2305 2022-05-11 08:07:19 |

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.
Zhu, S.; , . The Role of Cigarette Smoking on Bone Metabolism. Encyclopedia. Available online: https://encyclopedia.pub/entry/22703 (accessed on 18 May 2024).
Zhu S,  . The Role of Cigarette Smoking on Bone Metabolism. Encyclopedia. Available at: https://encyclopedia.pub/entry/22703. Accessed May 18, 2024.
Zhu, Sheng, . "The Role of Cigarette Smoking on Bone Metabolism" Encyclopedia, https://encyclopedia.pub/entry/22703 (accessed May 18, 2024).
Zhu, S., & , . (2022, May 09). The Role of Cigarette Smoking on Bone Metabolism. In Encyclopedia. https://encyclopedia.pub/entry/22703
Zhu, Sheng and . "The Role of Cigarette Smoking on Bone Metabolism." Encyclopedia. Web. 09 May, 2022.
The Role of Cigarette Smoking on Bone Metabolism
Edit

Cigarette smoking (CS) is recognized as an independent risk factor for the development of osteoporosis. Clinical studies have illustrated that smokers have significantly lower bone mineral density (BMD) than non-smokers, and cumulative bone loss can increase their lifetime risk of hip fracture by 50%. It has been shown that long-term CS can lead to an imbalance of bone turnover, further contributing to the reduction in bone mass and bone length and increased risk of fractures. Furthermore, chronic consumption of cigarettes has been increasingly linked to impaired muscle function.

cigarette smoking osteoporosis AhR pathway

1. Indirect Mechanisms

1.1. Body Weight

It has been shown that body weight is negatively associated with long-term CS [1][2][3]. This is probably because nicotine, a major addictive substance in cigarettes, inhibits food ingestion by stimulating the secretion of both dopamine and serotonin [4]. Additionally, CS has been shown to enhance lipid oxidation, contributing to the reduction in body weight [5]. In 2007, Wong et al. provided the underlying mechanisms explaining why long-term CS resulted in a reduction in body weight: (i) weight loss results in a reduction in the mechanical loading on bone, contributing to a decreased stimulus for osteogenic differentiation; (ii) CS cause a reduction in adipose tissue by inhibiting the conversation of androgen to estrogen; and (iii) CS cause a reduction in leptin levels, which has been known to enhance bone mass [6].

1.2. Parathyroid Hormone- (PTH-) Vitamin D Axis

The PTH-vitamin D axis exerts crucial functions in maintaining bone density and calcium hemostasis [7]. PTH has a regulatory function in regulating serum calcium levels by bone and kidney reabsorption, while vitamin D can modulate the absorption of calcium from the intestine [8][9]. Several studies have indicated that smoking downregulates vitamin D serum levels [10][11] and inhibits the production of PTH [12]. However, the influence of CS on the inhibition of the production of PTH remains controversial [13][14]. These inconsistencies may be due to confounding effects of body weight, alcohol consumption, medication use and calcium and vitamin D supplementation [10][15].

1.3. Gonadal Hormones

Estrogen and testosterone exert essential functions in modulating bone remodeling [16][17]. It has been shown that estrogen inhibits osteoclast differentiation and bone resorption [18], while testosterone promotes proliferation and osteogenic differentiation [19]. In females, CS causes a reduction in estrogen levels by altering estrogen metabolism [20][21]. Several explanations behind this phenomenon are as follows: (i) nicotine and cotinine suppress aromatase enzyme activity, contributing to a reduction in estrogen levels [22]; (ii) CS promotes estradiol to decompose into 2-methoxyestrone in the liver [23]; and (iii) CS enhances serum hormone-binding globulin levels, leading to a reduction in free estradiol levels in the blood [24][25]. In males, current findings are contradictory. Some research indicated testosterone levels were comparable between smokers and nonsmokers, whereas other research showed that smokers had higher testosterone levels than nonsmokers [26][27]. Like females, the potential mechanism of aromatase inhibition was involved in males [12].

1.4. Oxidative Stress

The presence of reactive oxygen species (ROS) in bone can influence resident cell behavior, extra-cellular matrix composition and tissue architecture. CS is related to high levels of ROS, which improves osteoclast activity and suppresses osteoblast activity, leading to a reduction in bone mass [28][29], and maqui berry extract, a fruit extract rich in anthocyanins, is effective against the effects of CSE on osteoblasts. A clinical study demonstrated that smoking increased levels of oxidative stress [30]. Consistently, Zhu and colleagues observed that cigarette smoke extract (CSE) significantly enhanced ROS and nitrotyrosine levels in human osteoblasts [31]. Probing potential molecular mechanisms, Aspera-Werz et al.reported that Nrf2 directly modulates antioxidant defense systems in human mesenchymal stem cells exposed to CSE [32], and resveratrol could decrease ROS level generated by CSE to protect the primary cilia integrity in human bone mesenchymal stem cells.

2. Direct Mechanisms

CS has been found to exert a direct role in bone tissue through binding to several receptors, including nicotinic acetylcholine receptors and androgen receptors in osteoblasts and aryl hydrocarbon receptors in osteoclasts [33]. Nicotine, a major addictive substance in cigarettes, binds to nicotinic receptors in osteoblasts [34]. However, the role of nicotine in bone metabolism has been controversial. Marinucci et al. observed that nicotine negatively contributed to the impaired osteogenic differentiation of mesenchymal stem cells (MSCs) [35]. On the contrary, Daffner and colleagues demonstrated that nicotine could even promote MSC proliferation and osteogenic differentiation at concentrations lower than its serum concentration in smokers [36]. Aspera-Werz et al. showed that the osteogenic differentiation of MSCs was not largely altered when the cells were exposed to nicotine at concentrations similar to its serum levels in smokers [32]. Similarly, Shintcovsk and colleagues demonstrated that nicotine had no influence on osteoclastogenesis [37]. Therefore, nicotine may not be a major compound responsible for impaired bone metabolism in bone cells.
Many studies have reported the negative relationship between CS and bone mass [38][39][40]. However, early diagnosis of impaired bone metabolism remains a great challenge. Due to the lack of obvious symptoms in the early stages of CS-induced osteoporosis, the bone metabolism of smokers has been impaired for a long period of time when they start to feel back pain or suffer from fractures [41][42]. Actually, after reaching peak bone mass around the age of 30 in adults, bone loss occurs at varying rates, and smoking significantly accelerates this process. Hence, it is highly desirable to detect the alternations of bone metabolism in the early stages to prevent the development of osteoporosis [43].
Several factors secreted into the blood by osteoblasts and osteoclasts regulate osteoblast-medicated bone formation and osteoclast-mediated bone resorption [44][45]. These serum markers represent a crucial complementary diagnostic tool to bone mineral density [46]. In general, monitoring these secreted markers could be helpful in determining the alternations in bone metabolism before the manifestation of osteoporosis. Figure 1 summarizes the direct effects of CS on bone metabolism.
Figure 1. Established gene markers for bone metabolism in (A) physiological conditions and (B) CS exposure conditions. Bone resorption markers include: tartrate-resistant acid phosphatase isoform 5b (TRAP5b), carbonic anhydrate II (CA II), C-terminal telopeptide (CTX) and N-terminal telopeptide (NTX). Regulators of osteoclastogenesis include: receptor activator of nuclear factor kappa B ligand (RANKL) and osteoprotegerin (OPG). Bone formation markers include: osteocalcin (OC), collagen I, bone-specific alkaline phosphatase (BAP), hydroxyproline (HYP) and type I collagen N- and C-terminal propeptides (PINP and PICP/CICP). Dotted arrows represent expression. Red arrows represent altered expression (up or down).

2.1. Markers for Bone Formation

Hydroxyproline (HYP) and type I collagen N- and C-terminal propeptides (PINP and PICP/CICP) in blood are commonly used to evaluate bone formation [47][48][49]. Ehnert and colleagues demonstrated that following total joint replacement, heavy smokers had a greater risk of prolonged hospital stay due to reduced levels of CICP as compared to nonsmokers [50]. Additionally, there are alternative markers involved in osteogenesis. Gürlek et al. reported that smoking significantly decreased salivary levels of osteocalcin (OC, later stage of osteogenic differentiation) [51]. Supporting these data, in a rat model, Gao et al. reported that chronic exposure to smoking significantly decreases levels of bone-specific alkaline phosphatase (BAP, early stage of osteogenic differentiation) and OC, resulting in osteoporotic bone [52].

2.2. Markers for Bone Degradation

C-terminal telopeptide (CTX) and N-terminal telopeptide (NTX) in blood are commonly used to represent collagen-I degradation [48][49]. Oncken et al. demonstrated that smoking cessation contributed to a reduction in NTX levels in postmenopausal women [53]. In addition, there are alternative markers involved in osteoblastogensis. An in vitro study revealed that chronic exposure to CSE-enhanced carbonic anhydrate II (CA II, early stage of osteoclastic differentiation [54]) and tartrate-resistant acid phosphatase 5b (TRAP 5b, later stage of osteoclastic differentiation [55]) activities in a bone co-culture system, resulting in an osteoporotic microenvironment [56].
Although these markers can represent bone metabolic alternations, the underlying mechanisms by which CS induces an osteoporotic bone environment are not yet fully understood. Understanding the regulatory mechanisms in bone cells impaired by CS will enable researchers to obtain insights into the development of CS-induced osteoporotic bones and screen relevant preventive treatment strategies for smokers with osteoporotic bones.

2.3. RANKL-RANK-OPG Pathway

It has been shown that the receptor activator of nuclear factor-kappa B ligand (RANKL)-RANK-osteoprotegerin (OPG) (RANKL-RANK-OPG) pathway exerts a crucial role in osteoclast differentiation and bone resorption [57][58][59]. Leibbrandt and colleagues revealed that osteoclasts failed to be differentiated in a RANKL and RANK knockout mouse model, indicating RANKL is crucial for osteoclastic differentiation and activation [60]. RANKL, a membrane protein, is synthesized and released by osteoblasts, which is capable of binding to the RANK receptor located on osteoclasts, activating osteoclastogenesis [61]. As a decoy receptor for RANKL, OPG suppresses osteoclastogenesis by binding to RANKL [62]. Hence, the balance between RANKL and OPG exerts a vital role in regulating bone metabolism [63].
Currently, few studies have been conducted regarding the association between CS and the RNKL-RANK-OPG pathway. In terms of human studies, researchers have demonstrated that smoking significantly reduced OPG levels, leading to an increased RANKL/OPG ratio [13][64]. Consistently, in an in vitro bone co-culture system, a research showed that chronic exposure to CSE enhanced osteoclast function via upregulation of the RANKL/OPG ratio, resulting in an osteoporotic microenvironment, and bisphosphonates can be used to counteract the effects of CSE on the RANKL/OPG pathway [56].
Several studies have shown the RANKL-RANK-OPG pathway is also involved in indirect pathophysiological mechanisms (PTH-vitamin D axis and gonadal hormones), which makes this pathway of great therapeutic interest [60]. Moreover, this pathway, in interaction with several factors, such as prostaglandin E2 and interleukins, further affects bone metabolism [60]. For instance, estrogen and androgen could enhance OPG levels in osteoblasts and downregulate the RANKL/OPG ratio, further inhibiting osteoclast differentiation [65].

2.4. Wnt/β-catenin Pathway

The canonical Wnt/β-catenin signaling pathway may be one potential mechanism that is involved in the regulation of RANKL/OPG balance. Glass et al. reported that the activation of the Wnt/β-catenin pathway in osteoblasts can downregulate the RANKL/OPG ratio, leading to compromised osteoclast differentiation and bone resorption [66]. Apart from suppressing osteoclastogenesis, it also enhances the osteogenic differentiation of MSCs [67], bone formation [68] and mineralization [69]. Therefore, the Wnt/β-catenin pathway appears to exert a dual role in bone remodeling, enhancing osteogenesis on the one hand and suppressing osteoclastogenesis on the other hand. Understanding and characterizing the Wnt/β-catenin pathway in detail is believed to be critical to guiding the interaction between osteoblast and osteoclast [70].
Wnt ligands, which secrete glycoproteins, are engaged in activating the Wnt/β-catenin canonical pathway [71]. When Wnt ligands (Wnt 3a and 5a) bind with receptor complexes, including Frizzled and LRP5/6, the Wnt/β-catenin pathway is activated to accumulate β-catenin in the cytoplasm of osteoblasts [72]. Then, the accumulated β-catenin transfers into the nucleus to regulate the transcription of osteogenic-related genes such as Osterix, RUNX2 and OPG [73]. Eventually, the accumulated OPG binds with RANKL and prevents it from integrating with RANKL located on osteoclasts, inhibiting osteoclastogenesis [74]. Additionally, sclerostin and Dickkopf-1 (DKK1) are potent targets, blocking the canonical Wnt signaling pathway [75]. DKK1 is expressed by osteocytes and mature osteoblast, while sclerostin is expressed by osteocytes [76].
To date, few studies have been carried out showing the association between CS and inhibitors of Wnt signaling. Jorde et al. reported that smokers had higher levels of DKK1 and lower levels of bone formation (PINP) when compared to nonsmokers [7]. Supporting these data, Miranda et al. revealed that smoking could enhance the levels of DKK1 and sclerostin in patients with periodontitis [77]. Thus, the inhibition of Wnt signaling by CS may result in the upregulation of the RANKL/OPG ratio, leading to excessive osteoclast differentiation and bone resorption (Figure 2).
Figure 2. Proposed regulatory mechanisms for bone metabolism in (A) physiological conditions and (B) smoking exposure conditions. Dotted arrows represent expression. Red arrows represent altered expression (up or down).

2.5. Aryl Hydrocarbon Receptor (AhR) Pathway

Smoking exposure potently activates the cellular Aryl hydrocarbon receptor (AhR) [78], as cigarette smoke contains a large number of exogenous ligands for AhR, such as dioxin-like derivatives represented by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and a variety of polycyclic aromatic hydrocarbon (PAH)-based compounds such as benzo(a)pyrene (BaP) [79]. The AhR is one of the ligand-activated transcription factors for sensing environmental pollutants, which stays in an inactive form in the cytoplasm. Upon binding to exogenous ligands from smoke, the conformation of the AhR is changed, which enables AhR translocation into the nucleus, where it heterodimerizes with the aromatic hydrocarbon receptor nuclear transfer protein (ARNT) and activates xenobiotic response elements (XRE), to the initial transcription of target genes (CYP1a1/CYP1a2/CYP1b1) and leads to different toxicological and pathological effects [79][80]. Recent studies have shown that endogenous activation of the AhR has multiple roles in the regulation of bone remodeling and its associated signaling pathways, which may be an essential mechanism for smoking-induced osteoporosis [81].
Jameel et al. reported that BaP and TCDD in cigarette smoke caused AhR activation and a significant increase in CYP1A1 and CYP1A2 expression, resulting in enhanced OC differentiation and compromised bone mass in wild-type mice but not in AhR-/- mice [33]. Moreover, TCDD administration was unable to stimulate osteoclastogenesis in bone marrow macrophages (BMMs) from CYP1A1/CYP1A2 double knockout and CYP1A1/CYP1A2/CYP1B1 triple knockout mice, unlike in BMM from wild-type mice [33]. These results suggest that AhR-induced CYP1 expression mediates at least in part the activation of osteoclastogenesis by AhR ligands from cigarette smoke. Further studies found that RANKL-stimulated osteoclastogenic signaling (e.g., phosphorylation of Akt, MAPK and NF-κB) was impaired in AhR-/- BMMs [82]. Furthermore, the AhR was found to be activated by RANKL at an earlier stage than signature osteoclastic genes (e.g., CSTK and NFATc1) in pre-osteoclasts, suggesting a regulatory role of the AhR in OC differentiation. BaP was shown to induce higher levels of c-Fos in RANKL-stimulated BMMs, but c-Fos was not induced by BaP or RANKL in AhR-/- BMMs [82]. Consistently, the AhR activation in BMMs was also found to lead to significant upregulation of c-Fos and NFATc1 expression, thereby initiating osteoclastic differentiation [83]. Taken together, the AhR pathway mediates excessive osteoclastic differentiation and bone resorption caused by smoking exposure, which could be a potential target for the prevention of smoking-induced osteoporosis (Figure 3).

Figure 3. Smoking exposure induces osteoclast differentiation through the AhR pathway, leading to osteoporosis. Dotted lines represent release, arrows represent promotion.

References

  1. Wang, G.Z.; Zhang, L.; Zhao, X.C.; Gao, S.H.; Qu, L.W.; Yu, H.; Fang, W.F.; Zhou, Y.C.; Liang, F.; Zhang, C.; et al. The Aryl hydrocarbon receptor mediates tobacco-induced PD-L1 expression and is associated with response to immunotherapy. Nat. Commun. 2019, 10, 1125.
  2. Xue, J.; Zhao, Q.; Sharma, V.; Nguyen, L.P.; Lee, Y.N.; Pham, K.L.; Edderkaoui, M.; Pandol, S.J.; Park, W.; Habtezion, A. Aryl Hydrocarbon Receptor Ligands in Cigarette Smoke Induce Production of Interleukin-22 to Promote Pancreatic Fibrosis in Models of Chronic Pancreatitis. Gastroenterology 2016, 151, 1206–1217.
  3. Gutiérrez-Vázquez, C.; Quintana, F.J. Regulation of the Immune Response by the Aryl Hydrocarbon Receptor. Immunity 2018, 48, 19–33.
  4. Yu, H.; Jiang, L.; Wan, B.; Zhang, W.; Yao, L.; Che, T.; Gan, C.; Su, N.; He, J.; Huang, J.; et al. The role of aryl hydrocarbon receptor in bone remodeling. Prog. Biophys. Mol. Biol. 2018, 134, 44–49.
  5. Iqbal, J.; Sun, L.; Cao, J.; Yuen, T.; Lu, P.; Bab, I.; Leu, N.A.; Srinivasan, S.; Wagage, S.; Hunter, C.A.; et al. Smoke carcinogens cause bone loss through the aryl hydrocarbon receptor and induction of Cyp1 enzymes. Proc. Natl. Acad. Sci. USA 2013, 110, 11115–11120.
  6. Izawa, T.; Arakaki, R.; Mori, H.; Tsunematsu, T.; Kudo, Y.; Tanaka, E.; Ishimaru, N. The Nuclear Receptor AhR Controls Bone Homeostasis by Regulating Osteoclast Differentiation via the RANK/c-Fos Signaling Axis. J. Immunol. 2016, 197, 4639–4650.
  7. Eisa, N.H.; Reddy, S.V.; Elmansi, A.M.; Kondrikova, G.; Kondrikov, D.; Shi, X.M.; Novince, C.M.; Hamrick, M.W.; McGee-Lawrence, M.E.; Isales, C.M.; et al. Kynurenine Promotes RANKL-Induced Osteoclastogenesis In Vitro by Activating the Aryl Hydrocarbon Receptor Pathway. Int. J. Mol. Sci. 2020, 21, 7931.
  8. Lips, P. Vitamin D physiology. Prog. Biophys. Mol. Biol. 2006, 92, 4–8.
  9. Khazai, N.; Judd, S.E.; Tangpricha, V. Calcium and vitamin D: Skeletal and extraskeletal health. Curr. Rheumatol. Rep. 2008, 10, 110–117.
  10. Brot, C.; Jorgensen, N.R.; Sorensen, O.H. The influence of smoking on vitamin D status and calcium metabolism. Eur. J. Clin. Nutr. 1999, 53, 920–926.
  11. Need, A.G.; Kemp, A.; Giles, N.; Morris, H.A.; Horowitz, M.; Nordin, B.E. Relationships between intestinal calcium absorption, serum vitamin D metabolites and smoking in postmenopausal women. Osteoporos. Int. 2002, 13, 83–88.
  12. Yoon, V.; Maalouf, N.M.; Sakhaee, K. The effects of smoking on bone metabolism. Osteoporos. Int. 2012, 23, 2081–2092.
  13. Al-Bashaireh, A.M.; Alqudah, O. Comparison of Bone Turnover Markers between Young Adult Male Smokers and Nonsmokers. Cureus 2020, 12, e6782.
  14. Fujiyoshi, A.; Polgreen, L.E.; Gross, M.D.; Reis, J.P.; Sidney, S.; Jacobs, D.R., Jr. Smoking habits and parathyroid hormone concentrations in young adults: The CARDIA study. Bone Rep. 2016, 5, 104–109.
  15. Kiel, D.P.; Zhang, Y.; Hannan, M.T.; Anderson, J.J.; Baron, J.A.; Felson, D.T. The effect of smoking at different life stages on bone mineral density in elderly men and women. Osteoporos. Int. 1996, 6, 240–248.
  16. Prestwood, K.M.; Pilbeam, C.C.; Burleson, J.A.; Woodiel, F.N.; Delmas, P.D.; Deftos, L.J.; Raisz, L.G. The short-term effects of conjugated estrogen on bone turnover in older women. J. Clin. Endocrinol. Metab. 1994, 79, 366–371.
  17. Mellström, D.; Johnell, O.; Ljunggren, O.; Eriksson, A.L.; Lorentzon, M.; Mallmin, H.; Holmberg, A.; Redlund-Johnell, I.; Orwoll, E.; Ohlsson, C. Free testosterone is an independent predictor of BMD and prevalent fractures in elderly men: MrOS Sweden. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 2006, 21, 529–535.
  18. Shevde, N.K.; Bendixen, A.C.; Dienger, K.M.; Pike, J.W. Estrogens suppress RANK ligand-induced osteoclast differentiation via a stromal cell independent mechanism involving c-Jun repression. Proc. Natl. Acad. Sci. USA 2000, 97, 7829–7834.
  19. Chen, Q.; Kaji, H.; Kanatani, M.; Sugimoto, T.; Chihara, K. Testosterone increases osteoprotegerin mRNA expression in mouse osteoblast cells. Horm. Metab. Res. 2004, 36, 674–678.
  20. Bjarnason, N.H.; Christiansen, C. The influence of thinness and smoking on bone loss and response to hormone replacement therapy in early postmenopausal women. J. Clin. Endocrinol. Metab. 2000, 85, 590–596.
  21. Michnovicz, J.J.; Hershcopf, R.J.; Naganuma, H.; Bradlow, H.L.; Fishman, J. Increased 2-hydroxylation of estradiol as a possible mechanism for the anti-estrogenic effect of cigarette smoking. N. Engl. J. Med. 1986, 315, 1305–1309.
  22. Barbieri, R.L.; Gochberg, J.; Ryan, K.J. Nicotine, cotinine, and anabasine inhibit aromatase in human trophoblast in vitro. J. Clin. Investig. 1986, 77, 1727–1733.
  23. Michnovicz, J.J.; Hershcopf, R.J.; Haley, N.J.; Bradlow, H.L.; Fishman, J. Cigarette smoking alters hepatic estrogen metabolism in men: Implications for atherosclerosis. Metabolism 1989, 38, 537–541.
  24. Cassidenti, D.L.; Vijod, A.G.; Vijod, M.A.; Stanczyk, F.Z.; Lobo, R.A. Short-term effects of smoking on the pharmacokinetic profiles of micronized estradiol in postmenopausal women. Am. J. Obs. Gynecol. 1990, 163, 1953–1960.
  25. Daniel, M.; Martin, A.D.; Drinkwater, D.T. Cigarette smoking, steroid hormones, and bone mineral density in young women. Calcif. Tissue Int. 1992, 50, 300–305.
  26. English, K.M.; Pugh, P.J.; Parry, H.; Scutt, N.E.; Channer, K.S.; Jones, T.H. Effect of cigarette smoking on levels of bioavailable testosterone in healthy men. Clin. Sci. 2001, 100, 661–665.
  27. Wang, W.; Yang, X.; Liang, J.; Liao, M.; Zhang, H.; Qin, X.; Mo, L.; Lv, W.; Mo, Z. Cigarette smoking has a positive and independent effect on testosterone levels. Hormones 2013, 12, 567–577.
  28. Valavanidis, A.; Vlachogianni, T.; Fiotakis, K. Tobacco smoke: Involvement of reactive oxygen species and stable free radicals in mechanisms of oxidative damage, carcinogenesis and synergistic effects with other respirable particles. Int. J. Environ. Res. Public Health 2009, 6, 445–462.
  29. Callaway, D.A.; Jiang, J.X. Reactive oxygen species and oxidative stress in osteoclastogenesis, skeletal aging and bone diseases. J. Bone Min. Metab. 2015, 33, 359–370.
  30. Çetin, A.; Muhtaro, S.; Saraymen, R.; Öztürk, A.; Muderris, I.J. Smoking-Induced Bone Defects May Be Due to Oxidative Damage in Postmenopausal Women. Turk. Klin. Tip Bilimleri Derg. 2009, 29, 851–858.
  31. Zhu, S.; Aspera-Werz, R.H.; Chen, T.; Weng, W.; Braun, B.; Histing, T.; Nüssler, A.K. Maqui berry extract prevents cigarette smoke induced oxidative stress in human osteoblasts in vitro. EXCLI J. 2021, 20, 281–296.
  32. Aspera-Werz, R.H.; Ehnert, S.; Heid, D.; Zhu, S.; Chen, T.; Braun, B.; Sreekumar, V.; Arnscheidt, C.; Nussler, A.K. Nicotine and Cotinine Inhibit Catalase and Glutathione Reductase Activity Contributing to the Impaired Osteogenesis of SCP-1 Cells Exposed to Cigarette Smoke. Oxid. Med. Cell. Longev. 2018, 2018, 3172480.
  33. Cusano, N.E. Skeletal Effects of Smoking. Curr. Osteoporos. Rep. 2015, 13, 302–309.
  34. Kim, B.S.; Kim, S.J.; Kim, H.J.; Lee, S.J.; Park, Y.J.; Lee, J.; You, H.K. Effects of nicotine on proliferation and osteoblast differentiation in human alveolar bone marrow-derived mesenchymal stem cells. Life Sci. 2012, 90, 109–115.
  35. Marinucci, L.; Balloni, S.; Fettucciari, K.; Bodo, M.; Talesa, V.N.; Antognelli, C. Nicotine induces apoptosis in human osteoblasts via a novel mechanism driven by H2O2 and entailing Glyoxalase 1-dependent MG-H1 accumulation leading to TG2-mediated NF-kB desensitization: Implication for smokers-related osteoporosis. Free Radic. Biol. Med. 2018, 117, 6–17.
  36. Daffner, S.D.; Waugh, S.; Norman, T.L.; Mukherjee, N.; France, J.C. Nicotine Increases Osteoblast Activity of Induced Bone Marrow Stromal Cells in a Dose-Dependent Manner: An in vitro Cell Culture Experiment. Glob. Spine J. 2012, 2, 153–158.
  37. Shintcovsk, R.L.; Knop, L.; Tanaka, O.M.; Maruo, H. Nicotine effect on bone remodeling during orthodontic tooth movement: Histological study in rats. Dent. Press J. Orthod. 2014, 19, 96–107.
  38. Hagiwara, S.; Tsumura, K. Smoking as a risk factor for bone mineral density in the heel of Japanese men. J. Clin. Densitom. 1999, 2, 219–222.
  39. Law, M.R.; Cheng, R.; Hackshaw, A.K.; Allaway, S.; Hale, A.K. Cigarette smoking, sex hormones and bone density in women. Eur. J. Epidemiol. 1997, 13, 553–558.
  40. Rudäng, R.; Darelid, A.; Nilsson, M.; Nilsson, S.; Mellström, D.; Ohlsson, C.; Lorentzon, M. Smoking is associated with impaired bone mass development in young adult men: A 5-year longitudinal study. J. Bone Miner. Res. 2012, 27, 2189–2197.
  41. Ulivieri, F.M.; Silva, B.C.; Sardanelli, F.; Hans, D.; Bilezikian, J.P.; Caudarella, R. Utility of the trabecular bone score (TBS) in secondary osteoporosis. Endocrine 2014, 47, 435–448.
  42. Sheu, A.; Diamond, T. Diagnostic tests: Bone mineral density: Testing for osteoporosis. Aust. Prescr. 2016, 39, 35.
  43. Christenson, E.S.; Jiang, X.; Kagan, R.; Schnatz, P. Osteoporosis management in post-menopausal women. Minerva Ginecol. 2012, 64, 181–194.
  44. Katagiri, T.; Takahashi, N. Regulatory mechanisms of osteoblast and osteoclast differentiation. Oral Dis. 2002, 8, 147–159.
  45. Matsuo, K.; Irie, N. Osteoclast–osteoblast communication. Arch. Biochem. Biophys. 2008, 473, 201–209.
  46. Shetty, S.; Kapoor, N.; Bondu, J.D.; Thomas, N.; Paul, T.V. Bone turnover markers: Emerging tool in the management of osteoporosis. Indian J. Endocrinol. Metab. 2016, 20, 846–852.
  47. Coates, P. Bone turnover markers. Aust. Fam. Physician. 2013, 42, 285–287.
  48. Nishizawa, Y.; Ohta, H.; Miura, M.; Inaba, M.; Ichimura, S.; Shiraki, M.; Takada, J.; Chaki, O.; Hagino, H.; Fujiwara, S.; et al. Guidelines for the use of bone metabolic markers in the diagnosis and treatment of osteoporosis (2012 edition). J. Bone Miner. Metab. 2013, 31, 1–15.
  49. Szulc, P. The role of bone turnover markers in monitoring treatment in postmenopausal osteoporosis. Clin. Biochem. 2012, 45, 907–919.
  50. Ehnert, S.; Aspera-Werz, R.H.; Ihle, C.; Trost, M.; Zirn, B.; Flesch, I.; Schröter, S.; Relja, B.; Nussler, A.K. Smoking dependent alterations in bone formation and inflammation represent major risk factors for complications following total joint arthroplasty. J. Clin. Med. 2019, 8, 406.
  51. Gurlek, O.; Lappin, D.F.; Buduneli, N. Effects of smoking on salivary C-telopeptide pyridinoline cross-links of type I collagen and osteocalcin levels. Arch. Oral. Biol. 2009, 54, 1099–1104.
  52. Gao, S.G.; Li, K.H.; Xu, M.; Jiang, W.; Shen, H.; Luo, W.; Xu, W.S.; Tian, J.; Lei, G.H. Bone turnover in passive smoking female rat: Relationships to change in bone mineral density. BMC Musculoskelet. Disord. 2011, 12, 131.
  53. Oncken, C.; Prestwood, K.; Cooney, J.L.; Unson, C.; Fall, P.; Kulldorff, M.; Raisz, L.G. Effects of smoking cessation or reduction on hormone profiles and bone turnover in postmenopausal women. Nicotine Tob. Res. 2002, 4, 451–458.
  54. Lehenkari, P.; Hentunen, T.A.; Laitala-Leinonen, T.; Tuukkanen, J.; Väänänen, H.K. Carbonic anhydrase II plays a major role in osteoclast differentiation and bone resorption by effecting the steady state intracellular pH and Ca2+. Exp. Cell Res. 1998, 242, 128–137.
  55. Jeong, J.W.; Choi, S.H.; Han, M.H.; Kim, G.Y.; Park, C.; Hong, S.H.; Lee, B.J.; Park, E.K.; Kim, S.O.; Leem, S.H.; et al. Protective Effects of Fermented Oyster Extract against RANKL-Induced Osteoclastogenesis through Scavenging ROS Generation in RAW 264.7 Cells. Int. J. Mol. Sci. 2019, 20, 1439.
  56. Zhu, S.; Haussling, V.; Aspera-Werz, R.H.; Chen, T.; Braun, B.; Weng, W.; Histing, T.; Nussler, A.K. Bisphosphonates Reduce Smoking-Induced Osteoporotic-Like Alterations by Regulating RANKL/OPG in an Osteoblast and Osteoclast Co-Culture Model. Int. J. Mol. Sci. 2020, 22, 53.
  57. Boyce, B.F.; Xing, L. The RANKL/RANK/OPG pathway. Curr. Osteoporos. Rep. 2007, 5, 98–104.
  58. Tobeiha, M.; Moghadasian, M.H.; Amin, N.; Jafarnejad, S. RANKL/RANK/OPG Pathway: A Mechanism Involved in Exercise-Induced Bone Remodeling. BioMed Res. Int. 2020, 2020, 6910312.
  59. Roshandel, D.; Holliday, K.L.; Pye, S.R.; Boonen, S.; Borghs, H.; Vanderschueren, D.; Huhtaniemi, I.T.; Adams, J.E.; Ward, K.A.; Bartfai, G.; et al. Genetic variation in the RANKL/RANK/OPG signaling pathway is associated with bone turnover and bone mineral density in men. J. Bone Min. Res. 2010, 25, 1830–1838.
  60. Leibbrandt, A.; Penninger, J.M. RANK/RANKL: Regulators of immune responses and bone physiology. Ann. N Y Acad. Sci. 2008, 1143, 123–150.
  61. Wada, T.; Nakashima, T.; Hiroshi, N.; Penninger, J.M. RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol. Med. 2006, 12, 17–25.
  62. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast differentiation by RANKL and OPG signaling pathways. J. Bone Miner. Metab. 2021, 39, 19–26.
  63. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch Biochem. Biophys. 2008, 473, 139–146.
  64. Lappin, D.F.; Sherrabeh, S.; Jenkins, W.M.; Macpherson, L.M. Effect of smoking on serum RANKL and OPG in sex, age and clinically matched supportive-therapy periodontitis patients. J. Clin. Periodontol. 2007, 34, 271–277.
  65. Syed, F.; Khosla, S. Mechanisms of sex steroid effects on bone. Biochem. Biophys. Res. Commun. 2005, 328, 688–696.
  66. Glass, D.A., 2nd; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev. Cell. 2005, 8, 751–764.
  67. Imamura, A.; Kajiya, H.; Fujisaki, S.; Maeshiba, M.; Yanagi, T.; Kojima, H.; Ohno, J. Three-dimensional spheroids of mesenchymal stem/stromal cells promote osteogenesis by activating stemness and Wnt/β-catenin. Biochem. Biophys. Res. Commun. 2020, 523, 458–464.
  68. Carrillo-Lopez, N.; Martinez-Arias, L.; Fernandez-Villabrille, S.; Ruiz-Torres, M.P.; Dusso, A.; Cannata-Andia, J.B.; Naves-Diaz, M.; Panizo, S.; European Renal Osteodystrophy, W. Role of the RANK/RANKL/OPG and Wnt/β-Catenin Systems in CKD Bone and Cardiovascular Disorders. Calcif. Tissue Int. 2021, 108, 439–451.
  69. Ruan, Y.; Kato, H.; Taguchi, Y.; Yamauchi, N.; Umeda, M. Irradiation by high-intensity red light-emitting diode enhances human bone marrow mesenchymal stem cells osteogenic differentiation and mineralization through Wnt/β-catenin signaling pathway. Lasers Med. Sci. 2021, 36, 55–65.
  70. Choi, H.K.; Kim, G.J.; Yoo, H.S.; Song, D.H.; Chung, K.H.; Lee, K.J.; Koo, Y.T.; An, J.H. Vitamin C Activates Osteoblastogenesis and Inhibits Osteoclastogenesis via Wnt/β-Catenin/ATF4 Signaling Pathways. Nutrients 2019, 11, 506.
  71. Zhang, Z.; Pan, X.; Chen, M.; Bai, M. Wnt signalling in oral and maxillofacial diseases. Cell Biol. Int. 2022, 46, 34–45.
  72. Wagner, J.M.; Steubing, Y.; Dadras, M.; Wallner, C.; Lotzien, S.; Huber, J.; Sogorski, A.; Sacher, M.; Reinkemeier, F.; Dittfeld, S.; et al. Wnt3a and ASCs are capable of restoring mineralization in staph aureus-infected primary murine osteoblasts. J. Bone Min. Metab. 2022, 40, 20–28.
  73. Xu, X.J.; Shen, L.; Yang, Y.P.; Zhu, R.; Shuai, B.; Li, C.G.; Wu, M.X. Serum β -Catenin Levels Associated with the Ratio of RANKL/OPG in Patients with Postmenopausal Osteoporosis. Int. J. Endocrinol. 2013, 2013, 534352.
  74. Chu, M.; Sun, Z.; Fan, Z.; Yu, D.; Mao, Y.; Guo, Y. Bi-directional regulation functions of lanthanum-substituted layered double hydroxide nanohybrid scaffolds via activating osteogenesis and inhibiting osteoclastogenesis for osteoporotic bone regeneration. Theranostics 2021, 11, 6717–6734.
  75. Evenepoel, P.; D’haese, P.; Brandenburg, V.J. Sclerostin and DKK1: New players in renal bone and vascular disease. Kidney Int. 2015, 88, 235–240.
  76. Moysés, R.M.; Schiavi, S.C. Sclerostin, osteocytes, and chronic kidney disease–mineral bone disorder. Semin. Dial. 2015, 28, 578–586.
  77. Miranda, T.S.; Napimoga, M.H.; Feres, M.; Marins, L.M.; da Cruz, D.F.; da Silva, H.D.P.; Duarte, P.M. Antagonists of Wnt/β-catenin signalling in the periodontitis associated with type 2 diabetes and smoking. J. Clin. Periodontol. 2018, 45, 293–302.
More
Information
Subjects: Orthopedics
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 618
Revisions: 4 times (View History)
Update Date: 11 May 2022
1000/1000