Assessment of tobacco heating system 2.4 on osteogenic differentiation of mesenchymal stem cells and primary human osteoblasts compared to conventional cigarettes

Romina H Aspera-Werz, Sabrina Ehnert, Monja Müller, Sheng Zhu, Tao Chen, Weidong Weng, Johann Jacoby, Andreas K Nussler, Romina H Aspera-Werz, Sabrina Ehnert, Monja Müller, Sheng Zhu, Tao Chen, Weidong Weng, Johann Jacoby, Andreas K Nussler

Abstract

Background: Cigarette smoking (CS) is the most common method of consuming tobacco. Deleterious effects on bone integrity, increased incidence of fractures, and delayed fracture healing are all associated with CS. Over 150 of the 6500 molecular species contained in cigarette smoke and identified as toxic compounds are inhaled by CS and, via the bloodstream, reach the skeletal system. New technologies designed to develop a reduced-risk alternative for smokers are based on electronic nicotine delivery systems, such as e-cigarettes and tobacco heating systems (THS). THS are designed to heat tobacco instead of burning it, thereby reducing the levels of harmful toxic compounds released.

Aim: To examine the effects of THS on osteoprogenitor cell viability and function compared to conventional CS.

Methods: Human immortalized mesenchymal stem cells (n = 3) and primary human pre-osteoblasts isolated from cancellous bone samples from BG Unfall Klinik Tübingen (n = 5) were osteogenically differentiated in vitro with aqueous extracts generated from either the THS 2.4 "IQOS" or conventional "Marlboro" cigarettes for up to 21 d. Cell viability was analyzed using resazurin conversion assay (mitochondrial activity) and calcein-AM staining (esterase activity). Osteogenic differentiation and bone cell function were evaluated using alkaline phosphatase (AP) activity, while matrix formation was analyzed through alizarin red staining. Primary cilia structure was examined by acetylated α-tubulin immunofluorescent staining. Free radical production was evaluated with 2',7'-dichlorofluorescein-diacetate assay.

Results: Our data clearly show that THS is significantly less toxic to bone cells than CS when analyzed by mitochondrial and esterase activity (P < 0.001). No significant differences in cytotoxicity between the diverse flavors of THS were observed. Harmful effects from THS on bone cell function were observed only at very high, non-physiological concentrations. In contrast, extracts from conventional cigarettes significantly reduced the AP activity (by two-fold) and matrix mineralization (four-fold) at low concentrations. Additionally, morphologic analysis of primary cilia revealed no significant changes in the length of the organelle involved in osteogenesis of osteoprogenitor cells, nor in the number of ciliated cells following THS treatment. Assessment of free radical production demonstrated that THS induced significantly less oxidative stress than conventional CS in osteoprogenitor cells.

Conclusion: THS was significantly less harmful to osteoprogenitor cells during osteogenesis than conventional CS. Additional studies are required to confirm whether THS is a better alternative for smokers to improve delays in bone healing following fracture.

Keywords: Bone; Cigarette smoke; Electronic nicotine delivery systems; Mesenchymal stem cells; Primary human osteoblast; Tobacco heating system.

Conflict of interest statement

Conflict-of-interest statement: The authors have not conflict of interest.

©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.

Figures

Figure 1
Figure 1
Tobacco heating systems and conventional cigarette aqueous extract characterization. A: Time to consume one unit [cigarette or tobacco heating systems (THS) stick] measured in minutes; B: pH from the aqueous extract (AE) generated with conventional cigarettes or THS; C: Absorbance at 320 nm from the different fractions produced from conventional cigarette or THS; D: Representative picture of gas washing bottle after conventional cigarette or THS AE generation, respectively. Each measure was conducted with six independent AE for each condition in triplicates. Data were analyzed using the nonparametric Mann Whitney test or the Kruskal-Wallis H test followed by Dunn’s post-hoc tests. Data are presented as mean ± SE, and P < 0.001 for the comparison. THS: Tobacco heating systems.
Figure 2
Figure 2
High concentrations of aqueous extract from tobacco heating systems has minor effects on bone cells viability after a single exposure than aqueous extract from conventional cigarettes. SCP-1 cells and primary human osteoblasts were treated with increasing concentrations of aqueous extract (AE) from conventional cigarettes and tobacco heating systems. Cell viability was evaluated by resazurin conversion (mitochondrial activity) in SCP-1 cells (A) and primary osteoblast (B) after 48 h of treatment; C: Representative live staining pictures from SCP-1 cells using calcein-AM (green) and nuclear staining using Hoechst 33342 (blue) was shown after 48 h of exposure to AE (scale bar 400 µm). Each measure was conducted at least three independent times in triplicates. Data were analyzed using the Kruskal-Wallis H test followed by Dunn’s post-hoc tests. Data are presented as mean ± SE, and P values indicated as bP < 0.01 and dP < 0.001 for comparisons with untreated cells within the same AE type. AE: Aqueous extract.
Figure 3
Figure 3
Tobacco heating systems is less toxic than conventional cigarettes after chronic exposure. SCP-1 cells and primary human osteoblasts were osteogenically differentiated with increasing concentrations of aqueous extract (AE) from conventional cigarette and tobacco heating systems (THS) for 21 d. Cell viability was evaluated by resazurin conversion (mitochondrial activity) in SCP-1 cells (A, B) and primary human osteoblast (C, D) after 14 (A, C) and 21 d (B, D). Each measurement was conducted at least three independent times in triplicates. Data were analyzed using the Kruskal-Wallis H test followed by Dunn’s post-hoc test. Data are presented as mean ± SE. P values are classified as aP < 0.05, bP < 0.01, cP < 0.001 for comparisons with untreated cells within AE type and as eP < 0.05, fP < 0.01, gP < 0.001 for comparisons of conventional cigarette with THS within the same concentration. AE: Aqueous extract.
Figure 4
Figure 4
Tobacco heating systems has a lower impact on bone cells function than conventional cigarettes. SCP-1 cells and primary human osteoblasts were osteogenically differentiated with increasing concentrations of aqueous extract (AE) from conventional cigarettes and tobacco heating systems (THS) for 21 d. Cell function was evaluated by alkaline phosphatase activity (early differentiation marker) (A, C) after 14 d. Calcium deposition (late marker of differentiation) was evaluated by Alizarin red staining (B, D) after 21 d (B, D). Each measurement was conducted at least three independent times in triplicates. Data were analyzed using the Kruskal-Wallis H test followed by Dunn’s post-hoc test. Data are represented as mean ± SE, and P values are classified as aP < 0.05, bP < 0.01, cP < 0.001 for comparisons with untreated cells within the same AE type and fP < 0.01, gP < 0.001 for comparisons conventional cigarette with THS within the same concentration. AE: Aqueous extract.
Figure 5
Figure 5
Tobacco heating systems induces less oxidative stress and causes less damage to primary cilia structures on bone cells precursor than conventional cigarettes. SCP-1 cells were treated with increasing concentrations of aqueous extract (AE) from conventional cigarettes and tobacco heating systems (THS). ROS production was evaluated by DCFH-DA assay in SCP-1 cells. 0.01%v/v H2O2 was used as a positive control (A). Primary cilium length was quantified on day 14 by acetylated α-tubulin (green), and nuclei (blue) immunostaining (B). Representative immunostaining images of SCP-1 cells primary cilia after 14 days of chronic exposure to THS or conventional cigarettes AE (C) (scale bar 100 µm). Each measurement was conducted at least three independent times in triplicates. Data were analyzed using the Kruskal-Wallis H test followed by Dunn’s post-hoc tests. Data are represented as mean ± SE, and P values are classified as aP < 0.05, bP < 0.01, cP < 0.001 for comparisons with untreated cells within AE type. AE: Aqueous extract.
Figure 6
Figure 6
No differential variation in cytotoxicity between different tobacco heating systems flavors on bone cells precursor. SCP-1 cells were treated with increasing concentrations of aqueous extract (AE) from tobacco heating systems bronce, amber and yellow flavor. Cell cytotoxicity was evaluated by resazurin conversion (mitochondrial activity) in SCP-1 cells after 48 h of exposure. Each measurement was conducted at least three independent times in triplicates. Data were analyzed using the Kruskal-Wallis H test followed by Dunn’s post-hoc tests. Data are presented as mean ± SE, and P values are classified as bP < 0.01, cP < 0.001 for comparisons with untreated cells within AE type. AE: Aqueous extract.

References

    1. Kahnert S, Pötschke-Langer M, Kahnert S, Viarisio V, Heidt C, Schunk S, Mons U, Fode K. 2015. Tabakatlas deutschland 2015. Pabst Science Publishers.
    1. GBD 2015 Tobacco Collaborators. Smoking prevalence and attributable disease burden in 195 countries and territories, 1990-2015: a systematic analysis from the Global Burden of Disease Study 2015. Lancet. 2017;389:1885–1906.
    1. Cooke M. The chemical components of tobacco and tobacco smoke. Chromatographia. 2010;71:977–977.
    1. Cusano NE. Skeletal Effects of Smoking. Curr Osteoporos Rep. 2015;13:302–309.
    1. Wong PK, Christie JJ, Wark JD. The effects of smoking on bone health. Clin Sci (Lond) 2007;113:233–241.
    1. Ward KD, Klesges RC. A meta-analysis of the effects of cigarette smoking on bone mineral density. Calcif Tissue Int. 2001;68:259–270.
    1. Kanis JA, Johnell O, Oden A, Johansson H, De Laet C, Eisman JA, Fujiwara S, Kroger H, McCloskey EV, Mellstrom D, Melton LJ, Pols H, Reeve J, Silman A, Tenenhouse A. Smoking and fracture risk: a meta-analysis. Osteoporos Int. 2005;16:155–162.
    1. Amin S, Niu J, Guermazi A, Grigoryan M, Hunter DJ, Clancy M, LaValley MP, Genant HK, Felson DT. Cigarette smoking and the risk for cartilage loss and knee pain in men with knee osteoarthritis. Ann Rheum Dis. 2007;66:18–22.
    1. Adams CI, Keating JF, Court-Brown CM. Cigarette smoking and open tibial fractures. Injury. 2001;32:61–65.
    1. Scolaro JA, Schenker ML, Yannascoli S, Baldwin K, Mehta S, Ahn J. Cigarette smoking increases complications following fracture: a systematic review. J Bone Joint Surg Am. 2014;96:674–681.
    1. Mills E, Eyawo O, Lockhart I, Kelly S, Wu P, Ebbert JO. Smoking cessation reduces postoperative complications: a systematic review and meta-analysis. Am J Med. 2011;124:144–154.e8.
    1. Abate M, Vanni D, Pantalone A, Salini V. Cigarette smoking and musculoskeletal disorders. Muscles Ligaments Tendons J. 2013;3:63–69.
    1. Singh JA, Schleck C, Harmsen WS, Jacob AK, Warner DO, Lewallen DG. Current tobacco use is associated with higher rates of implant revision and deep infection after total hip or knee arthroplasty: A prospective cohort study. BMC Med. 2015;13:283.
    1. Ehnert S, Aspera-Werz RH, Ihle C, Trost M, Zirn B, Flesch I, Schroter S, Relja B, Nussler AK. Smoking dependent alterations in bone formation and inflammation represent major risk factors for complications following total joint arthroplasty. J Clin Med. 2019;8:406.
    1. Rothem DE, Rothem L, Soudry M, Dahan A, Eliakim R. Nicotine modulates bone metabolism-associated gene expression in osteoblast cells. J Bone Miner Metab. 2009;27:555–561.
    1. Pappas RS. Toxic elements in tobacco and in cigarette smoke: inflammation and sensitization. Metallomics. 2011;3:1181–1198.
    1. U.S. Food and Drug Administration. Harmful and potentially harmful constituents in tobacco products and tobacco smoke: Established list. 2012. Available from: .
    1. Aspera-Werz RH, Ehnert S, Heid D, Zhu S, Chen T, Braun B, Sreekumar V, Arnscheidt C, Nussler AK. 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.
    1. Daffner SD, Waugh S, Norman TL, Mukherjee N, France JC. Nicotine Increases Osteoblast Activity of Induced Bone Marrow Stromal Cells in a Dose-Dependent Manner: An in vitro Cell Culture Experiment. Global Spine J. 2012;2:153–158.
    1. Kim BS, Kim SJ, Kim HJ, Lee SJ, Park YJ, Lee J, You HK. Effects of nicotine on proliferation and osteoblast differentiation in human alveolar bone marrow-derived mesenchymal stem cells. Life Sci. 2012;90:109–115.
    1. Braun KF, Ehnert S, Freude T, Egaña JT, Schenck TL, Buchholz A, Schmitt A, Siebenlist S, Schyschka L, Neumaier M, Stöckle U, Nussler AK. Quercetin protects primary human osteoblasts exposed to cigarette smoke through activation of the antioxidative enzymes HO-1 and SOD-1. ScientificWorldJournal. 2011;11:2348–2357.
    1. Ehnert S, Braun KF, Buchholz A, Freude T, Egaña JT, Schenck TL, Schyschka L, Neumaier M, Döbele S, Stöckle U, Nussler AK. Diallyl-disulphide is the effective ingredient of garlic oil that protects primary human osteoblasts from damage due to cigarette smoke. Food Chem. 2012;132:724–729.
    1. Ehnert S, Stefan D, Friedrich BK, Britta B, Valeska H, Mario H, Tomas EJ, Ulrich S, Thomas F, Klaus NA. N-acetylcyteine and flavonoid rich diet: The protective effect of 15 different antioxidants on cigarette smoke-damaged primary human osteoblasts. Adv Biosci Biotechnol. 2012;3:1129–1139.
    1. Holzer N, Braun KF, Ehnert S, Egaña JT, Schenck TL, Buchholz A, Schyschka L, Neumaier M, Benzing S, Stöckle U, Freude T, Nussler AK. Green tea protects human osteoblasts from cigarette smoke-induced injury: possible clinical implication. Langenbecks Arch Surg. 2012;397:467–474.
    1. Sreekumar V, Aspera-Werz R, Ehnert S, Strobel J, Tendulkar G, Heid D, Schreiner A, Arnscheidt C, Nussler AK. Resveratrol protects primary cilia integrity of human mesenchymal stem cells from cigarette smoke to improve osteogenic differentiation in vitro. Arch Toxicol. 2018;92:1525–1538.
    1. Centers for Disease Control and Prevention (CDC) Quitting smoking among adults--United States, 2001-2010. MMWR Morb Mortal Wkly Rep. 2011;60:1513–1519.
    1. Haziza C, de La Bourdonnaye G, Donelli A, Poux V, Skiada D, Weitkunat R, Baker G, Picavet P, Lüdicke F. Reduction in Exposure to Selected Harmful and Potentially Harmful Constituents Approaching Those Observed Upon Smoking Abstinence in Smokers Switching to the Menthol Tobacco Heating System 2.2 for 3 Months (Part 1) Nicotine Tob Res. 2020;22:539–548.
    1. Li X, Luo Y, Jiang X, Zhang H, Zhu F, Hu S, Hou H, Hu Q, Pang Y. Chemical Analysis and Simulated Pyrolysis of Tobacco Heating System 2.2 Compared to Conventional Cigarettes. Nicotine Tob Res. 2019;21:111–118.
    1. Margham J, McAdam K, Forster M, Liu C, Wright C, Mariner D, Proctor C. Chemical Composition of Aerosol from an E-Cigarette: A Quantitative Comparison with Cigarette Smoke. Chem Res Toxicol. 2016;29:1662–1678.
    1. Davis B, To V, Talbot P. Comparison of cytotoxicity of IQOS aerosols to smoke from Marlboro Red and 3R4F reference cigarettes. Toxicol In Vitro. 2019;61:104652.
    1. Phillips B, Szostak J, Titz B, Schlage WK, Guedj E, Leroy P, Vuillaume G, Martin F, Buettner A, Elamin A, Sewer A, Sierro N, Choukrallah MA, Schneider T, Ivanov NV, Teng C, Tung CK, Lim WT, Yeo YS, Vanscheeuwijck P, Peitsch MC, Hoeng J. A six-month systems toxicology inhalation/cessation study in ApoE-/- mice to investigate cardiovascular and respiratory exposure effects of modified risk tobacco products, CHTP 1.2 and THS 2.2, compared with conventional cigarettes. Food Chem Toxicol. 2019;126:113–141.
    1. Munakata S, Ishimori K, Kitamura N, Ishikawa S, Takanami Y, Ito S. Oxidative stress responses in human bronchial epithelial cells exposed to cigarette smoke and vapor from tobacco- and nicotine-containing products. Regul Toxicol Pharmacol. 2018;99:122–128.
    1. Bozier J, Zakarya R, Chapman DG, Oliver BGG. How harmless are E-cigarettes? Effects in the pulmonary system. Curr Opin Pulm Med. 2020;26:97–102.
    1. Gotts JE, Jordt SE, McConnell R, Tarran R. What are the respiratory effects of e-cigarettes? BMJ. 2019;366:l5275.
    1. Ehnert S, Linnemann C, Aspera-Werz RH, Bykova D, Biermann S, Fecht L, De Zwart PM, Nussler AK, Stuby F. Immune Cell Induced Migration of Osteoprogenitor Cells Is Mediated by TGF-β Dependent Upregulation of NOX4 and Activation of Focal Adhesion Kinase. Int J Mol Sci. 2018;19
    1. Böcker W, Yin Z, Drosse I, Haasters F, Rossmann O, Wierer M, Popov C, Locher M, Mutschler W, Docheva D, Schieker M. Introducing a single-cell-derived human mesenchymal stem cell line expressing hTERT after lentiviral gene transfer. J Cell Mol Med. 2008;12:1347–1359.
    1. ISO and health canada intense smoking parameters - Part 2: Examination of factors contributing to variability in the routine measurement of TPM, water and NFDPM smoke yields of cigarettes. ISO, 2015. Available from: .
    1. Häussling V, Deninger S, Vidoni L, Rinderknecht H, Ruoß M, Arnscheidt C, Athanasopulu K, Kemkemer R, Nussler AK, Ehnert S. Impact of Four Protein Additives in Cryogels on Osteogenic Differentiation of Adipose-Derived Mesenchymal Stem Cells. Bioengineering (Basel) 2019;6
    1. Aspera-Werz RH, Chen T, Ehnert S, Zhu S, Fröhlich T, Nussler AK. Cigarette Smoke Induces the Risk of Metabolic Bone Diseases: Transforming Growth Factor Beta Signaling Impairment via Dysfunctional Primary Cilia Affects Migration, Proliferation, and Differentiation of Human Mesenchymal Stem Cells. Int J Mol Sci. 2019;20
    1. Ehnert S, Sreekumar V, Aspera-Werz RH, Sajadian SO, Wintermeyer E, Sandmann GH, Bahrs C, Hengstler JG, Godoy P, Nussler AK. TGF-β1 impairs mechanosensation of human osteoblasts via HDAC6-mediated shortening and distortion of primary cilia. J Mol Med (Berl) 2017;95:653–663.
    1. Huang W, Yang S, Shao J, Li YP. Signaling and transcriptional regulation in osteoblast commitment and differentiation. Front Biosci. 2007;12:3068–3092.
    1. Tummala P, Arnsdorf EJ, Jacobs CR. The Role of Primary Cilia in Mesenchymal Stem Cell Differentiation: A Pivotal Switch in Guiding Lineage Commitment. Cell Mol Bioeng. 2010;3:207–212.
    1. Anderson CT, Castillo AB, Brugmann SA, Helms JA, Jacobs CR, Stearns T. Primary cilia: cellular sensors for the skeleton. Anat Rec (Hoboken) 2008;291:1074–1078.
    1. Kode A, Rajendrasozhan S, Caito S, Yang SR, Megson IL, Rahman I. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2008;294:L478–L488.
    1. Sewer A, Kogel U, Talikka M, Wong ET, Martin F, Xiang Y, Guedj E, Ivanov NV, Hoeng J, Peitsch MC. Evaluation of the Tobacco Heating System 2.2 (THS2.2). Part 5: microRNA expression from a 90-day rat inhalation study indicates that exposure to THS2.2 aerosol causes reduced effects on lung tissue compared with cigarette smoke. Regul Toxicol Pharmacol. 2016;81 Suppl 2:S82–S92.
    1. Haswell LE, Corke S, Verrastro I, Baxter A, Banerjee A, Adamson J, Jaunky T, Proctor C, Gaça M, Minet E. In vitro RNA-seq-based toxicogenomics assessment shows reduced biological effect of tobacco heating products when compared to cigarette smoke. Sci Rep. 2018;8:1145.
    1. James AW. Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation. Scientifica (Cairo) 2013;2013:684736.

Source: PubMed

3
Abonnere