iconOpen Access

MINI-REVIEW

crossmark

Effects of areca nut consumption on cell differentiation of osteoblasts, myoblasts, and fibroblasts

YUNG-FU CHANG1,2,3,*

1 Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
2 Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
3 Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan

* Corresponding Author: YUNG-FU CHANG. Email: email

(This article belongs to this Special Issue: Cellular and Molecular Toxicology in Reproductive and Developmental Biology)

BIOCELL 2023, 47(2), 283-287. https://doi.org/10.32604/biocell.2023.025743

Abstract

Areca nut is used worldwide as a hallucinogenic addicting drug along the tropical belt. Arecoline, a toxic compound, is the most important alkaloid in areca nuts. The adverse effects of oral uptake and chewing of areca nut are well known. For example, the possibility of cancer caused by chewing areca nuts is widely discussed. Chewing areca nut has other adverse effects on other organs, including abnormal cell differentiation, oral cancer, and several other diseases. The use of areca nut is also associated with low birthweight. Skeletal musculature is the largest organ in the body and is attached to the bones. During embryo development, the differentiation of bone and muscle cells is critical. In this article, we reviewed the effects of areca nut and arecoline on embryonic cell differentiation, particularly osteoblasts, myoblasts, and fibroblasts.

Keywords


Introduction

Areca or betel nut is used worldwide by 600 million people, with India and Southeast Asian countries being the major consumers of areca nut (Arora and Squier, 2019). Arecoline (methyl 1-methyl-3,6-dihydro-2H-pyridine-5-carboxylate) is the most important alkaloid and the main toxic compound in areca nuts (Chen et al., 2021). Most studies have examined the role of areca nut and arecoline on the pathogenesis of oral lesions (Das and Giri, 2020). However, areca nut also affects most organ systems, such as reproductive organs, heart, brain, gastrointestinal tract, and lungs. Areca nut consumption also increases the risk of cardiac arrhythmias, asthma, metabolic syndrome, myocardial infarction, neuronal injury, hepatotoxicity, type II diabetes, central obesity, and hyperlipidemia (Garg et al., 2014). The above-mentioned diseases, such as cardiac arrhythmias, asthma, and metabolic syndrome, involve dysregulated cell differentiation. Arrhythmogenic cardiomyopathy is characterized by myocardial dysfunction and fibrofatty replacement of myocardial tissue. It may result from abnormal differentiation of cardiac progenitor cells to adipocytes (Lombardi et al., 2011). Asthma is an inflammatory disease. The differentiation of Th2/Th17 cells is increasingly dysregulated and is associated with asthma severity (Vroman et al., 2015). Obesity is a risk factor for the development of metabolic disorders. Subcutaneous-preadipocyte differentiation is associated with metabolic syndrome (Park et al., 2012). Cancers are considered developmental disorders that disrupt the normal cell differentiation (Tiwari, 2012). Long-term chewing of betel nut causes sperm reduction, asthma, uterine, mouth, and esophageal cancers (Chen et al., 2021). A systematic review showed that areca nut chewing also increases the risk of developing liver diseases (Khasbage et al., 2022). The risk of oral cancer increases with the daily number of quids consumed and the number of years of chewing betel nut in a dose-responsive manner (Warnakulasuriya and Chen, 2022). A systematic review showed that the use of areca (betel nut) is significantly associated with low birthweight (de Silva et al., 2019); consumption of high doses of areca nut leads to an arrest of endothelial cell differentiation and subsequent dysfunction of the fetus (Al-Rmalli et al., 2011).

In this review, we focused on the effects of areca nut on cell differentiation.

Osteoblast cell differentiation

The skeletal muscle is the largest organ in the body (Pedersen, 2013). The main function of bone is to support the attachment of muscles. Bone remodeling is regulated by the balance between osteoclasts and osteoblasts (Kim et al., 2020). Decreased bone formation by osteoblasts and increased bone resorption by osteoclasts result in alveolar bone loss (Mori et al., 2007). Osteoclasts are differentiated from monocyte/macrophage lineage cells. Hematopoietic stem cells (HSCs) are capable of self-renewal and have the pluripotent ability to differentiate into all hematopoietic cell types (Yahara et al., 2020). DAPI (4′, 6-diamidino-2-phenylindole dihydrochloride) can be used to trace short periods of osteogenic differentiation of mesenchymal stem cells (Ocarino et al., 2008). HSCs give rise to oligopotent progenitor cells, such as common myeloid progenitor cells (CMPs), upon differentiation, which in turn, differentiate into macrophages and osteoclasts (Ono and Nakashima, 2018). Macrophage precursors differentiate into marked monocytes and then into tissue-specific macrophages. These tissue-specific macrophages can fuse to provide osteoclasts, or sometimes they fuse into multinucleated giant cells to further differentiate into osteoclasts (Yao et al., 2021). Multinucleated osteoclasts work for bone resorption as induced by the receptor activator of nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) (Suda et al., 1992).

RANKL is a key regulator in osteoclastogenesis. It plays indispensable and irreplaceable functions in the osteoclast differentiation program by stimulating several osteoclastogenic pathways, such as NF-κB, mitogen-activated protein kinases (MAPK), and immunoreceptor tyrosine-based activation motif signals, as well as suppressing inhibitory molecules, such as tumor necrosis factor receptor-associated factor 3, p100, retinol binding protein-J, interferon regulatory factor 8, MAF bZIP transcription factor B, and B-cell lymphoma 6 (Takayanagi, 2021). RANKL is a membrane-associated cytokine expressed by osteoblasts. Osteoclast precursors express RANK, a RANKL receptor, to recognize RANKL by cell-cell interaction and differentiate into osteoclasts. Inhibition of RANKL-RANK signaling results in increased bone mass by preventing osteoclastic-regulated bone resorption (Udagawa et al., 2021). Osteoclasts can be generated in vitro by inducing macrophage lineage cells with RANKL treatment (Kurotaki et al., 2020). When primary bone marrow macrophages were treated with CSF and RANKL, 50–100 μM arecoline reduced the development of multinucleated osteoclasts by suppressing the expression of osteoclast differentiation-related genes through interference with the AKT, MAPK, and NF-kB activation pathways (Liu et al., 2020). Ling et al. (2005) showed that ripe areca nut extracts (rANE) can modulate the expression of RANKL. The expression of RANKL mRNA and protein in osteoblast-like MG63 cells is stimulated by rANE in a dose-dependent manner. However, the viability of these cells was reduced with rANE treatment.

Bone marrow stromal cells can be differentiated into qsteoblast-like cells (Rodriguez et al., 2009). KUSA/A1 cells (bone marrow stromal cell line) differentiated into osteoblast-like cells and induced bone tissue by both in vitro cell culture and intra-abdominal diffusion chambers implanted in SCID mice.

Muscle cell differentiation

Arecoline exposure results in a decrease in locomotor activity caused by defective somatic skeletal muscle development and mitochondrial dysfunction. In arecoline-exposed zebrafish embryos, reduced locomotor activity and swimming ability impairment were observed (Peng et al., 2015). Immunofluorescent staining and ultrastructural observations revealed a defective arrangement of myosin heavy chains and abnormal myofibril arrangement; arecoline also inhibited the embryonic development of mice. Arecoline could decrease the number of implanted embryos in mice at early pregnancy (Liu et al., 2011) and also inhibit the expansion of trophoblast outgrowth of blastocysts. Taken together, arecoline was harmful to mouse embryos as early as the peri-implantation stage (Liu et al., 2011). C2C12, a myoblast cell line, is a good material to study skeletal muscle cell differentiation. However, high passage numbers induce resistance to apoptosis (Pronsato et al., 2013). High (>60) passage numbers of C2C12 cells were found depleted of mitochondrial DNA (mtDNA) and resistant to H2O2 induction of apoptosis. We used low (<20) passage numbers of C2C12 for fur studies. It was shown that arecoline could inhibit the myogenic C2C12 cell differentiation by reducing the activated signal transducer and activator of transcription 3 (STAT3). At 0.4 mM concentration of arecoline, apoptosis increased, and the viability of C2C12 cells decreased (Chang et al., 2012). Myogenic differentiation markers such as myosin heavy chain and myogenin were inhibited by arecoline. Furthermore, arecoline inhibited the activated form of phosphorylated STAT3 during myotube formation. The clustering of acetylcholine receptors (AChRs) at the neuromuscular junction (NMJ), initiated by the protein agrin was important for developing muscular function. It was demonstrated in this study that arecoline could inhibit the formation of agrin-induced AChR clusters as well as destabilize agrin-induction spontaneously. These results demonstrate the adverse effects of arecoline from areca nut on muscle development (Chang et al., 2013). Arecoline was reported to generate reactive oxygen species (ROS) as well as to induce apoptosis (Yen et al., 2011). Oxidative stress is important for the pathological development of skeletal musculature (Musarò et al., 2010). To prevent the damage caused by oxidative stress on differentiating myoblasts, N-acetyl-cysteine (NAC), a scavenger of ROS, is involved in the generation of hepatic glutathione (GSH) to remove drugs (Yan et al., 2018). It can decrease oxidative stress and glutathione-dependent damage in cells, such as rat pancreatic Rin-5F cells in the presence of high concentrations of glucose and fatty acids, and increase the viability of rat C6 astroglia-like cells treated with cocaine (Alnahdi et al., 2019; Badisa et al., 2015). Our recent study showed that when C2C12 cells were treated with arecoline, NAC regenerated a decreased number of myotubes as well as nuclei in each myotube (Li et al., 2022). In addition, there was a minor improvement in NAC-mediated expression of myogenin and MYH, two myogenic markers, caused by arecoline. MEK/ERK signaling was shown to participate in the maintenance of myogenic progenitor cells (Miyake et al., 2020). Knockdown of ERK1/2 inhibits C2C12 cell differentiation (Feng et al., 2013). Recently, one study showed that NAC could restore the arecoline-induced decrease in the expression of p-ERK1/2 (Li et al., 2022). Areca nut is significantly associated with low birthweight in human studies (de Silva et al., 2019; Senn et al., 2009).

Fibroblast differentiation

Fibrosis development is a response to inflammation and epithelial injury resulting in the accumulation of extracellular matrix and myofibroblast activation (Edeling et al., 2016). Myofibroblasts differentiating from fibroblasts and other cell types, contribute to tissue repair during wound healing. Differentiated myofibroblasts are characterized by increased production of extracellular matrix (ECM) proteins. However, they can impair organ function when the contraction and ECM protein secretion become excessive, such as in Dupuytren’s disease, scleroderma, hypertrophic scars, and heart and kidney fibrosis (Hinz et al., 2007). Betel quid chewing can increase the risk of oral submucous fibrosis OSF, a collagen deposition (Shen et al., 2020). Areca nuts contain the components considered the main causative factors in the OSF. The trauma caused by coarse areca nut fiber and alkaloids induces tissue inflammation, fibroblast proliferation, collagen deposition, and myofibroblast differentiation to finally develop into OSF and oral cancer. During these processes, ECM turnover changes. The rearrangement of ECM is regulated by molecules such as plasminogen activator inhibitor-1 (PAI-1), transforming growth factor (TGF)-β1, lysyl oxidase, cystatin, metalloproteinases, and tissue inhibitors of metalloproteinases (Cheng et al., 2020). Slug is an EMT inducer associated with fibrogenesis and carcinogenesis. Arecoline increases the expression of Slug in normal fibrotic buccal mucosal fibroblasts (fBMFs). The inhibition of Slug can prevent arecoline-induced myofibroblast activation. The binding of Slug to the E-box of type I collagen promoter enhances the expression of type I collagen. These results showed the importance of arecoline-induced fibrogenesis (Fang et al., 2019). The expression of microRNA (miR)-21 was induced by arecoline treatment in a dose-dependent manner in BMFs through TGF-β signaling. The myofibroblast characteristics, such as higher cell motility and collagen gel contractility, induced by arecoline were suppressed by the miR-21 inhibitor. Moreover, miR-21 and myofibroblast marker, smooth muscle actin-alpha (α-SMA), were positively correlated. Therefore, the overexpression of miR-21 may be caused by the activation of areca nut components through the TGF-β pathway (Yang et al., 2021). Areca nut water extract induced p-SMAD2, an effector of TGF-β signaling, and TGF-β downstream targets, such as transglutaminase 2 (TGM2), transmembrane prostate androgen-induced RNA (TMEPAI), thrombospondin 1, and TGF-β1) in human epithelial keratinocyte HaCaT cells. These results suggest the important role of TGF-β induced by areca nut in OSF progression (Khan et al., 2012). Profibrotic long noncoding RNAs (lncRNAs) H19 have been reported as overexpressed in several fibrotic diseases. With arecoline treatment, the expression of H19 was dose-dependently upregulated in BMFs. The results further demonstrated that arecoline stimulated the upregulation of H19 through the TGF-β pathway (Yu et al., 2021). Copper was reported in tissue fibrogenesis to cross-link collagen through the copper-dependent enzyme lysyl oxidase. The mean tissue copper level was higher in the OSF specimens compared with the non-areca chewing controls among patients with OSF. This finding suggests that copper is an initiating factor in OSF to stimulate fibrogenesis by increasing lysyl oxidase activity (Trivedy et al., 2000). TGF-β1 is the primary inducer of fibrosis and plays an important role in myofibroblast formation. The activation of myofibroblasts includes rapid synthesis of ECM components, such as collagen and fibronectin, during the repair of pathological tissues (Shu and Lovicu, 2017). Arecoline can induce wound healing capacities, invasion, migration, and collagen gel contractility of BMFs. Arctigenin, a lignan extracted from Arctium lappa, is capable of abolishing these myofibroblast characteristics of fBMFs through TGF-β/Smad signaling (Lin et al., 2019). Areca quid can cause OSF via ROS generation. TGM-2 is a matrix protein related to several fibrotic disorders and can be induced by ROS. Fibroblasts derived from OSF were found to have higher TGM-2 expression than normal BMFs. TGM-2 and ROS induced by arecoline were found in BMFs. These results suggest that arecoline induces ROS generation and upregulates TGM-2 expression, leading to ECM accumulation in OSF tissues of areca nut chewers (Lee et al., 2016). Hypoxia-inducible factor-1 (HIF-1) is a heterodimer containing α and β subunits as key mediators of cellular adaptation to low oxygen. It can regulate several profibrogenic genes related to tissue fibrosis. Arecoline was found capable of enhancing the expression of HIF-1α protein dose-dependently. PAI-1 is important for the inhibition of plasmin-dependent ECM degradation to accumulate ECM, and hypoxia can increase arecoline-induced PAI-1 protein expression. These data showed that the level of HIF-1α protein increased in OSF tissues from areca quid chewers (Tsai et al., 2015). Production of collagen of the ECM and induction of EMT is associated with renal cell fibrosis, and E-cadherin, N-cadherin, and vimentin are markers of EMT. TGF-β was shown to mediate the progression of renal fibrosis. Arecoline caused a dose-dependent decrease in E-cadherin expression and increases in α-SMA, vimentin, N-cadherin, and collagen in cultured human kidney (HK2) cells. In addition, arecoline also increases the expression of the phosphorylated extracellular signal-regulated kinase (ERK), suggesting that arecoline is important for inducing the EMT and fibrogenesis in renal tubule cells through ERK-mediated signaling pathways (Hsieh et al., 2020).

Conclusion

The adverse effects of areca nut have been the subject of several studies recently. In addition to oral diseases, areca nut also inhibits the differentiation of different cell types, such as osteoblasts and myoblasts, and promotes the fibrogenesis of fibroblasts. Areca nut is significantly associated with low birthweight in human studies. For embryonic development, the differentiation of bone and muscle cells is critical. Skeletal muscle is the largest organ in the body, and the primary function of the bones is to support the attachment of muscles; besides, muscle strength increases with the increase in muscle mass. These results will be helpful in understanding the mechanisms behind adverse effects, such as oral diseases, cancer progression, fibrosis of different organs, and ill effects on embryonic development caused by the consumption of areca nut (Fig. 1).

images

Figure 1: The effects of areca nut on cell differentiation. Consumption of areca nut can inhibit the differentiation of osteoclasts and myoblasts and stimulate fibrogenesis of fibroblasts.

Acknowledgement: I thank the constant support provided by Kaohsiung Medical University (Taiwan) for this study.

Author Contribution: The author confirms sole responsibility for the following: article conception and manuscript preparation.

Ethics Approval: Not applicable.

Funding Statement: I acknowledge the funding provided by the Ministry of Science and Technology, Taiwan (108-2314-B-037-075) and the Kaohsiung Medical University Research Foundation (KMU-M103001, KMU-M104003, KMU-TP104PR16).

Conflicts of Interest: The authors declare that they have no conflicts of interest to report regarding the present study.

References

  1. Al-Rmalli, SW., Jenkins, RO., & Haris, PI. (2011). Betel quid chewing elevates human exposure to arsenic, cadmium and lead. Journal of Hazardous Materials, 190, 69-74. [Google Scholar] [CrossRef]
  2. Alnahdi, A., John, A., & Raza, H. (2019). N-acetyl cysteine attenuates oxidative stress and glutathione-dependent redox imbalance caused by high glucose/high palmitic acid treatment in pancreatic Rin-5F cells. PLoS One, 14, e0226696. [Google Scholar] [CrossRef]
  3. Arora, S., & Squier, C. (2019). Areca nut trade, globalisation and its health impact: Perspectives from India and South-East Asia. Perspectives in Public Health, 139, 44-48. [Google Scholar] [CrossRef]
  4. Badisa, RB., Kumar, SS., Mazzio, E., Haughbrook, RD., Allen, JR., Davidson, MW., Fitch-Pye, CA., & Goodman, CB. (2015). N-acetyl cysteine mitigates the acute effects of cocaine-induced toxicity in astroglia-like cells. PLoS One, 10, e0114285. [Google Scholar] [CrossRef]
  5. Chang, YF., Liu, TY., & Liu, ST. (2013). Arecoline inhibits and destabilizes agrin-induced acetylcholine receptor cluster formation in C2C12 myotubes. Food and Chemical Toxicology, 60, 391-396. [Google Scholar] [CrossRef]
  6. Chang, YF., Liu, TY., Liu, ST., & Tseng, CN. (2012). Arecoline inhibits myogenic differentiation of C2C12 myoblasts by reducing STAT3 phosphorylation. Food and Chemical Toxicology, 50, 3433-3439. [Google Scholar] [CrossRef]
  7. Chen, X., He, Y., & Deng, Y. (2021). Chemical composition, pharmacological, and toxicological effects of betel nut. Evidence-Based Complementary and Alternative Medicine, 2021, 1808081. [Google Scholar] [CrossRef]
  8. Cheng, RH., Wang, YP., Chang, JY., Pan, YH., Chang, MC., & Jeng, JH. (2020). Genetic susceptibility and protein expression of extracellular matrix turnover-related genes in oral submucous fibrosis. International Journal of Molecular Sciences, 21, 8104. [Google Scholar] [CrossRef]
  9. Das, A., & Giri, S. (2020). A review on role of arecoline and its metabolites in the molecular pathogenesis of oral lesions with an insight into current status of its metabolomics. Prague Medical Report, 121, 209-235. [Google Scholar] [CrossRef]
  10. de Silva, M., Panisi, L., Brownfoot, FC., Lindquist, A., Walker, SP., Tong, S., & Hastie, R. (2019). Systematic review of areca (betel nut) use and adverse pregnancy outcomes. International Journal of Gynecology & Obstetrics, 147, 292-300. [Google Scholar] [CrossRef]
  11. Edeling, M., Ragi, G., Huang, S., Pavenstädt, H., & Susztak, K. (2016). Developmental signalling pathways in renal fibrosis: The roles of Notch, Wnt and Hedgehog. Nature Reviews Nephrology, 12, 426-439. [Google Scholar] [CrossRef]
  12. Fang, CY., Hsia, SM., Hsieh, PL., Liao, YW., Peng, CY., Wu, CZ., Lin, KC., Tsai, LL., & Yu, CC. (2019). Slug mediates myofibroblastic differentiation to promote fibrogenesis in buccal mucosa. Journal of Cellular Physiology, 234, 6721-6730. [Google Scholar] [CrossRef]
  13. Feng, Y., Niu, LL., Wei, W., Zhang, WY., Li, XY., Cao, JH., & Zhao, SH. (2013). A feedback circuit between miR-133 and the ERK1/2 pathway involving an exquisite mechanism for regulating myoblast proliferation and differentiation. Cell Death & Disease, 4, e934. [Google Scholar] [CrossRef]
  14. Garg, A., Chaturvedi, P., & Gupta, PC. (2014). A review of the systemic adverse effects of areca nut or betel nut. Indian Journal of Medical and Paediatric Oncology, 35, 3-9. [Google Scholar] [CrossRef]
  15. Hinz, B., Phan, SH., Thannickal, VJ., Galli, A., Bochaton-Piallat, ML., & Gabbiani, G. (2007). The myofibroblast: One function, multiple origins. The American Journal of Pathology, 170, 1807-1816. [Google Scholar] [CrossRef]
  16. Hsieh, YH., Syu, RJ., Lee, CC., Lin, SH., Lee, CH., Cheng, CW., & Tsai, JP. (2020). Arecoline induces epithelial mesenchymal transition in HK2 cells by upregulating the ERK-mediated signaling pathway. Environmental Toxicology, 35, 1007-1014. [Google Scholar] [CrossRef]
  17. Khan, I., Kumar, N., Pant, I., Narra, S., & Kondaiah, P. (2012). Activation of TGF-β pathway by areca nut constituents: A possible cause of oral submucous fibrosis. PLoS One, 7, e51806. [Google Scholar] [CrossRef]
  18. Khasbage, SBD., Bhowate, RR., & Khatib, N. (2022). Risk of liver disease in areca nut habitual: A systematic review. Journal of Oral and Maxillofacial Pathology, 26, 128-129. [Google Scholar] [CrossRef]
  19. Kim, JM., Lin, C., Stavre, Z., Greenblatt, MB., & Shim, JH. (2020). Osteoblast-osteoclast communication and bone homeostasis. Cells, 9, 2073. [Google Scholar] [CrossRef]
  20. Kurotaki, D., Yoshida, H., & Tamura, T. (2020). Epigenetic and transcriptional regulation of osteoclast differentiation. Bone, 138, 115471. [Google Scholar] [CrossRef]
  21. Lee, SS., Chen, YJ., Tsai, CH., Huang, FM., & Chang, YC. (2016). Elevated transglutaminase-2 expression mediates fibrosis in areca quid chewing-associated oral submucocal fibrosis via reactive oxygen species generation. Clinical Oral Investigations, 20, 1029-1034. [Google Scholar] [CrossRef]
  22. Li, YX., Hsiao, CH., & Chang, YF. (2022). N-acetyl cysteine prevents arecoline-inhibited C2C12 myoblast differentiation through ERK1/2 phosphorylation. PLoS One, 17, e0272231. [Google Scholar] [CrossRef]
  23. Lin, CY., Hsieh, PL., Liao, YW., Peng, CY., Yu, CC., & Lu, MY. (2019). Arctigenin reduces myofibroblast activities in oral submucous fibrosis by LINC00974 inhibition. International Journal of Molecular Sciences, 20, 1328. [Google Scholar] [CrossRef]
  24. Ling, LJ., Ho, FC., Chen, YT., Holborow, DW., Liu, TY., & Hung, SL. (2005). Areca nut extracts modulated expression of alkaline phosphatase and receptor activator of nuclear factor κB ligand in osteoblasts. Journal of Clinical Periodontology, 32, 353-359. [Google Scholar] [CrossRef]
  25. Liu, FL., Chen, CL., Lai, CC., Lee, CC., & Chang, DM. (2020). Arecoline suppresses RANKL-induced osteoclast differentiation and attenuates LPS-induced bone loss . Phytomedicine, 69, 153195. [Google Scholar] [CrossRef]
  26. Liu, ST., Young, GC., Lee, YC., & Chang, YF. (2011). A preliminary report on the toxicity of arecoline on early pregnancy in mice. Food and Chemical Toxicology, 49, 144-148. [Google Scholar] [CrossRef]
  27. Lombardi, R., da Graca Cabreira-Hansen, M., Bell, A., Fromm, RR., Willerson, JT., & Marian, AJ. (2011). Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy. Circulation Research, 109, 1342-1353. [Google Scholar] [CrossRef]
  28. Miyake, T., Aziz, A., & McDermott, JC. (2020). Maintenance of the undifferentiated state in myogenic progenitor cells by TGFβ signaling is smad independent and requires MEK activation. International Journal of Molecular Sciences, 21, 1057. [Google Scholar] [CrossRef]
  29. Mori, G., Brunetti, G., Colucci, S., Ciccolella, F., & Coricciati, M. (2007). Alteration of activity and survival of osteoblasts obtained from human periodontitis patients: Role of TRAIL. Journal of Biological Regulators and Homeostatic Agents, 21, 105-114. [Google Scholar]
  30. Musarò, A., Fulle, S., & Fanò, G. (2010). Oxidative stress and muscle homeostasis. Current Opinion in Clinical Nutrition and Metabolic Care, 13, 236-242. [Google Scholar] [CrossRef]
  31. Ocarino, NM., Bozzi, A., Pereira, RD., Breyner, NM., Silva, VL., Castanheira, P., Goes, AM., & Serakides, R. (2008). Behavior of mesenchymal stem cells stained with 4′, 6-diamidino-2-phenylindole dihydrochloride (DAPI) in osteogenic and non osteogenic cultures. BIOCELL, 32, 175-183. [Google Scholar] [CrossRef]
  32. Ono, T., & Nakashima, T. (2018). Recent advances in osteoclast biology. Histochemistry and Cell Biology, 149, 325-341. [Google Scholar] [CrossRef]
  33. Park, HT., Lee, ES., Cheon, YP., Lee, DR., Yang, KS., Kim, YT., Hur, JY., Kim, SH., Lee, KW., & Kim, T. (2012). The relationship between fat depot-specific preadipocyte differentiation and metabolic syndrome in obese women. Clinical Endocrinology, 76, 59-66. [Google Scholar] [CrossRef]
  34. Pedersen, BK. (2013). Muscle as a secretory organ. Comprehensive Physiology, 3, 1337-1362. [Google Scholar] [CrossRef]
  35. Peng, WH., Lee, YC., Chau, YP., Lu, KS., & Kung, HN. (2015). Short-term exposure of zebrafish embryos to arecoline leads to retarded growth, motor impairment, and somite muscle fiber changes. Zebrafish, 12, 58-70. [Google Scholar] [CrossRef]
  36. Pronsato, L., La Colla, A., Ronda, AC., Milanesi, L., Boland, R., & Vasconsuelo, A. (2013). High passage numbers induce resistance to apoptosis in C2C12 muscle cells. BIOCELL, 37, 1-9. [Google Scholar] [CrossRef]
  37. Rodriguez, AP., Tsujigiwa, H., Gunduz, M., Cengiz, B., Nagai, N., Tamamura, R., Borkosky, SS., Takagi, T., Inoue, M., & Nagatsuka, H. (2009). Influence of the microenvironment on gene and protein expression of odontogenic-like and osteogenic-like cells. BIOCELL, 33, 39-47. [Google Scholar] [CrossRef]
  38. Senn, M., Baiwog, F., Winmai, J., Mueller, I., Rogerson, S., & Senn, N. (2009). Betel nut chewing during pregnancy, Madang Province, Papua New Guinea. Drug and Alcohol Dependence, 105, 126-131. [Google Scholar] [CrossRef]
  39. Shen, YW., Shih, YH., Fuh, LJ., & Shieh, TM. (2020). Oral submucous fibrosis: A review on biomarkers, pathogenic mechanisms, and treatments. International Journal of Molecular Sciences, 21, 7231. [Google Scholar] [CrossRef]
  40. Shu, DY., & Lovicu, FJ. (2017). Myofibroblast transdifferentiation: The dark force in ocular wound healing and fibrosis. Progress in Retinal and Eye Research, 60, 44-65. [Google Scholar] [CrossRef]
  41. Suda, T., Takahashi, N., & Martin, TJ. (1992). Modulation of osteoclast differentiation. Endocrine Reviews, 13, 66-80. [Google Scholar] [CrossRef]
  42. Takayanagi, H. (2021). RANKL as the master regulator of osteoclast differentiation. Journal of Bone and Mineral Metabolism, 39, 13-18. [Google Scholar] [CrossRef]
  43. Tiwari, M. (2012). Microarrays and cancer diagnosis. Journal of Cancer Research and Therapeutics, 8, 3-10. [Google Scholar] [CrossRef]
  44. Trivedy, CR., Warnakulasuriya, KA., Peters, TJ., Senkus, R., Hazarey, VK., & Johnson, NW. (2000). Raised tissue copper levels in oral submucous fibrosis. Journal of Oral Pathology & Medicine, 29, 241-248. [Google Scholar] [CrossRef]
  45. Tsai, CH., Lee, SS., & Chang, YC. (2015). Hypoxic regulation of plasminogen activator inhibitor-1 expression in human buccal mucosa fibroblasts stimulated with arecoline. Journal of Oral Pathology & Medicine, 44, 669-673. [Google Scholar] [CrossRef]
  46. Udagawa, N., Koide, M., Nakamura, M., Nakamichi, Y., & Yamashita, T. (2021). Osteoclast differentiation by RANKL and OPG signaling pathways. Journal of Bone and Mineral Metabolism, 39, 19-26. [Google Scholar] [CrossRef]
  47. Vroman, H., van den Blink, B., & Kool, M. (2015). Mode of dendritic cell activation: The decisive hand in Th2/Th17 cell differentiation. Implications in asthma severity?. Immunobiology, 220, 254-261. [Google Scholar] [CrossRef]
  48. Warnakulasuriya, S., & Chen, THH. (2022). Areca nut and oral cancer: Evidence from studies conducted in humans. Journal of Dental Research, 101, 1139-1146. [Google Scholar] [CrossRef]
  49. Yahara, Y., Barrientos, T., Tang, YJ., Puviindran, V., & Nadesan, P. (2020). Erythromyeloid progenitors give rise to a population of osteoclasts that contribute to bone homeostasis and repair. Nature Cell Biology, 22, 49-59. [Google Scholar] [CrossRef]
  50. Yan, M., Huo, Y., Yin, S., & Hu, H. (2018). Mechanisms of acetaminophen-induced liver injury and its implications for therapeutic interventions. Redox Biology, 17, 274-283. [Google Scholar] [CrossRef]
  51. Yang, HW., Yu, CC., Hsieh, PL., Liao, YW., Chu, PM., Yu, CH., & Fang, CY. (2021). Arecoline enhances miR-21 to promote buccal mucosal fibroblasts activation. Journal of the Formosan Medical Association, 120, 1108-1113. [Google Scholar] [CrossRef]
  52. Yao, Y., Cai, X., Ren, F., Ye, Y., Wang, F., Zheng, C., Qian, Y., & Zhang, M. (2021). The macrophage-osteoclast axis in osteoimmunity and osteo-related diseases. Frontiers in Immunology, 12, 664871. [Google Scholar] [CrossRef]
  53. Yen, CY., Lin, MH., Liu, SY., Chiang, WF., Hsieh, WF., Cheng, YC., Hsu, KC., & Liu, YC. (2011). Arecoline-mediated inhibition of AMP-activated protein kinase through reactive oxygen species is required for apoptosis induction. Oral Oncology, 47, 345-351. [Google Scholar] [CrossRef]
  54. Yu, CC., Liao, YW., Hsieh, PL., & Chang, YC. (2021). Targeting lncRNA H19/miR-29b/COL1A1 axis impedes myofibroblast activities of precancerous oral submucous fibrosis. International Journal of Molecular Sciences, 22, 2216. [Google Scholar] [CrossRef]

Cite This Article

CHANG, Y. (2023). Effects of areca nut consumption on cell differentiation of osteoblasts, myoblasts, and fibroblasts. BIOCELL, 47(2), 283–287.


cc This work is licensed under a Creative Commons Attribution 4.0 International License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
  • 1104

    View

  • 411

    Download

  • 0

    Like

Share Link