J Bone Metab.  2018 Feb;25(1):43-51. 10.11005/jbm.2018.25.1.43.

The Effect of Antidepressants on Mesenchymal Stem Cell Differentiation

Affiliations
  • 1Sydney Medical School Nepean, The University of Sydney, Penrith, Australia. gustavo.duque@unimelb.edu.au
  • 2Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia.
  • 3Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, Australia.
  • 4Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, Australia.

Abstract

BACKGROUND
Use of antidepressant medications has been linked to detrimental impacts on bone mineral density and osteoporosis; however, the cellular basis behind these observations remains poorly understood. The effect does not appear to be homogeneous across the whole class of drugs and may be linked to affinity for the serotonin transporter system. In this study, we hypothesized that antidepressants have a class- and dose-dependent effect on mesenchymal stem cell (MSC) differentiation, which may affect bone metabolism.
METHODS
Human MSCs (hMSCs) were committed to differentiate when either adipogenic or osteogenic media was added, supplemented with five increasing concentrations of amitriptyline (0.001-10 µM), venlafaxine (0.01-25 µM), or fluoxetine (0.001-10 µM). Alizarin red staining (mineralization), alkaline phosphatase (osteoblastogenesis), and oil red O (adipogenesis) assays were performed at timed intervals. In addition, cell viability was assessed using a MTT.
RESULTS
We found that fluoxetine had a significant inhibitory effect on mineralization. Furthermore, adipogenic differentiation of hMSC was affected by the addition of amitriptyline, venlafaxine, and fluoxetine to the media. Finally, none of the tested medications significantly affected cell survival.
CONCLUSIONS
This study showed a divergent effect of three antidepressants on hMSC differentiation, which appears to be independent of class and dose. As fluoxetine and amitriptyline, but not venlafaxine, affected both osteoblastogenesis and adipogenesis, this inhibitory effect could be associated to the high affinity of fluoxetine to the serotonin transporter system.

Keyword

Adipogenesis; Antidepressive agents; Mesenchymal stromal cells; Osteoblasts; Osteoporosis

MeSH Terms

Adipogenesis
Alkaline Phosphatase
Amitriptyline
Antidepressive Agents*
Bone Density
Cell Survival
Fluoxetine
Humans
Mesenchymal Stromal Cells*
Metabolism
Miners
Osteoblasts
Osteoporosis
Serotonin Plasma Membrane Transport Proteins
Venlafaxine Hydrochloride
Alkaline Phosphatase
Amitriptyline
Antidepressive Agents
Fluoxetine
Serotonin Plasma Membrane Transport Proteins
Venlafaxine Hydrochloride

Figure

  • Fig. 1 Effect of antidepressants on mineralization. Human mesenchymal stem cells (MSCs) were induced to differentiate into osteoblasts for 3 weeks at 37℃ in the presence of various concentrations of (A, B) amitriptyline, (C, D) venlafaxine, or (E, F) fluoxetine. Differentiating cells were also incubated with vehicle (VEH) alone, while undifferentiated cells were incubated with MSC growth media (GM) for 3 weeks as controls. Cells were then fixed and stained with Alizarin red (AR) as described in the methods. Changes in absorbance were measured relative to VEH only control. The extent of AR staining was quantified by solubilizing the stain with cetylpyridinium chloride and measuring the absorbance. Ratios were obtained by comparing absorbances with that of VEH. (A, B) Co-incubation with amitriptyline showed a modest decrease in AR staining only at the highest concentration tested, 10 µM. (C, D) Whereas co-incubation with venlafaxine showed no significant change in staining, (E, F) the addition of fluoxetine decreased staining at all concentrations tested. Undifferentiated hMSC showed no AR staining (GM bars). Data are representative of 3 to 6 independent experiments per drug. Images were taken under ×10 magnification. *P<0.05 compared to VEH-treated cells.

  • Fig. 2 Effect of antidepressants on alkaline phosphatase activity. Alkaline phosphatase activity was measured in human mesenchymal stem cells (MSCs) that had been induced to differentiate into osteoblasts over 3 weeks at 37℃ in the presence of various concentrations of (A) amitriptyline, (B) venlafaxine, or (C) fluoxetine. Differentiating cells were also incubated with vehicle (VEH) alone, while undifferentiated cells were incubated with MSC growth media (GM) for 3 weeks as controls. Alkaline phosphatase activity is displayed relative to VEH control. Data are representative of 3 to 6 independent experiments per drug. *P<0.05 compared to VEH-treated cells.

  • Fig. 3 Effect of antidepressants on adipogenesis. Human mesenchymal stem cells were induced to differentiate into adipocytes for 2 weeks at 37℃ in the presence of various concentrations of (A, B) amitriptyline, (C, D) venlafaxine, or (E, F) fluoxetine. Differentiating cells were also incubated with vehicle (VEH), while undifferentiated cells were incubated with MSC growth media (GM) for 2 weeks as controls. Cells were then fixed, stained with oil red O (ORO) and counterstained with hematoxylin as described in the methods. The extent of staining was then quantified by solubilizing the stain as described in the methods. Intensity of staining was determined by absorbance relative to VEH. Data are representative of 3 to 6 independent experiments per drug. Images were taken under ×10 magnification. *P<0.05 compared to VEH-treated cells.

  • Fig. 4 Assessment of cell viability. Human mesenchymal stem cells (MSC) induced to differentiate into osteoblasts (alizarin red [AR]) or adipocytes (oil red O [ORO]) in the presence of various concentrations of amitriptyline (Ami), venlafaxine (Ven), or fluoxetine (Fl). The Figure shows the effect of increasing concentrations of the drugs on cell viability. No significant effect was found at any treated conditions. Cells cultured in MSC growth media (GM) were used as control. *P<0.05 compared to differentiation media. VEH, vehicle.


Reference

1. Black DM, Rosen CJ. Clinical practice. Postmenopausal osteoporosis. N Engl J Med. 2016; 374:254–262.
2. Valenti MT, Dalle Carbonare L, Mottes M. Osteogenic differentiation in healthy and pathological conditions. Int J Mol Sci. 2016; 18:41.
Article
3. Verma S, Rajaratnam JH, Denton J, et al. Adipocytic proportion of bone marrow is inversely related to bone formation in osteoporosis. J Clin Pathol. 2002; 55:693–698.
Article
4. Gimble JM, Nuttall ME. Bone and fat: old questions, new insights. Endocrine. 2004; 23:183–188.
Article
5. Bermeo S, Gunaratnam K, Duque G. Fat and bone interactions. Curr Osteoporos Rep. 2014; 12:235–242.
Article
6. James AW. Review of signaling pathways governing MSC osteogenic and adipogenic differentiation. Scientifica (Cairo). 2013; 2013:684736.
Article
7. Charbord P. Bone marrow mesenchymal stem cells: historical overview and concepts. Hum Gene Ther. 2010; 21:1045–1056.
Article
8. Jacobs SA, Roobrouck VD, Verfaillie CM, et al. Immunological characteristics of human mesenchymal stem cells and multipotent adult progenitor cells. Immunol Cell Biol. 2013; 91:32–39.
Article
9. Zhang Y, Khan D, Delling J, et al. Mechanisms underlying the osteo- and adipo-differentiation of human mesenchymal stem cells. ScientificWorldJournal. 2012; 2012:793823.
Article
10. Tang QQ, Lane MD. Adipogenesis: from stem cell to adipocyte. Annu Rev Biochem. 2012; 81:715–736.
Article
11. Steiner JA, Carneiro AM, Blakely RD. Going with the flow: trafficking-dependent and -independent regulation of serotonin transport. Traffic. 2008; 9:1393–1402.
Article
12. Monti JM, Jantos H. The roles of dopamine and serotonin, and of their receptors, in regulating sleep and waking. Prog Brain Res. 2008; 172:625–646.
Article
13. Filip M, Bader M. Overview on 5-HT receptors and their role in physiology and pathology of the central nervous system. Pharmacol Rep. 2009; 61:761–777.
Article
14. Geldenhuys WJ, Van der Schyf CJ. The serotonin 5-HT6 receptor: a viable drug target for treating cognitive deficits in Alzheimer's disease. Expert Rev Neurother. 2009; 9:1073–1085.
Article
15. Abrams JK, Johnson PL, Hollis JH, et al. Anatomic and functional topography of the dorsal raphe nucleus. Ann N Y Acad Sci. 2004; 1018:46–57.
Article
16. Hornung JP. The human raphe nuclei and the serotonergic system. J Chem Neuroanat. 2003; 26:331–343.
Article
17. Michelsen KA, Schmitz C, Steinbusch HW. The dorsal raphe nucleus-from silver stainings to a role in depression. Brain Res Rev. 2007; 55:329–342.
Article
18. Gustafsson BI, Thommesen L, Stunes AK, et al. Serotonin and fluoxetine modulate bone cell function in vitro. J Cell Biochem. 2006; 98:139–151.
Article
19. Dimitri P, Rosen C. The central nervous system and bone metabolism: an evolving story. Calcif Tissue Int. 2017; 100:476–485.
Article
20. Oh CM, Park S, Kim H. Serotonin as a new therapeutic target for diabetes mellitus and obesity. Diabetes Metab J. 2016; 40:89–98.
Article
21. Namkung J, Kim H, Park S. Peripheral serotonin: a new player in systemic energy homeostasis. Mol Cells. 2015; 38:1023–1028.
Article
22. Karsenty G, Yadav VK. Regulation of bone mass by serotonin: molecular biology and therapeutic implications. Annu Rev Med. 2011; 62:323–331.
Article
23. Tatsumi M, Groshan K, Blakely RD, et al. Pharmacological profile of antidepressants and related compounds at human monoamine transporters. Eur J Pharmacol. 1997; 340:249–258.
Article
24. Elhwuegi AS. Central monoamines and their role in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2004; 28:435–451.
Article
25. Holtzheimer PE 3rd, Nemeroff CB. Advances in the treatment of depression. NeuroRx. 2006; 3:42–56.
Article
26. Homberg JR, Schubert D, Gaspar P. New perspectives on the neurodevelopmental effects of SSRIs. Trends Pharmacol Sci. 2010; 31:60–65.
Article
27. Magni LR, Purgato M, Gastaldon C, et al. Fluoxetine versus other types of pharmacotherapy for depression. Cochrane Database Syst Rev. 2013; Cd004185.
Article
28. Owens MJ, Morgan WN, Plott SJ, et al. Neurotransmitter receptor and transporter binding profile of antidepressants and their metabolites. J Pharmacol Exp Ther. 1997; 283:1305–1322.
29. Li B, Zhang S, Zhang H, et al. Fluoxetine-mediated 5-HT2B receptor stimulation in astrocytes causes EGF receptor transactivation and ERK phosphorylation. Psychopharmacology (Berl). 2008; 201:443–458.
Article
30. Li B, Zhang S, Li M, et al. Chronic treatment of astrocytes with therapeutically relevant fluoxetine concentrations enhances cPLA2 expression secondary to 5-HT2B-induced, transactivation-mediated ERK1/2 phosphorylation. Psychopharmacology (Berl). 2009; 207:1–12.
Article
31. Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG, et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacology. 2001; 25:871–880.
Article
32. Wong SY, Lau EM, Lynn H, et al. Depression and bone mineral density: is there a relationship in elderly Asian men? Results from Mr. Os (Hong Kong). Osteoporos Int. 2005; 16:610–615.
Article
33. Silverman SL, Shen W, Minshall ME, et al. Prevalence of depressive symptoms in postmenopausal women with low bone mineral density and/or prevalent vertebral fracture: results from the Multiple Outcomes of Raloxifene Evaluation (MORE) study. J Rheumatol. 2007; 34:140–144.
34. Erez HB, Weller A, Vaisman N, et al. The relationship of depression, anxiety and stress with low bone mineral density in post-menopausal women. Arch Osteoporos. 2012; 7:247–255.
Article
35. Robbins J, Hirsch C, Whitmer R, et al. The association of bone mineral density and depression in an older population. J Am Geriatr Soc. 2001; 49:732–736.
Article
36. Diem SJ, Blackwell TL, Stone KL, et al. Depressive symptoms and rates of bone loss at the hip in older women. J Am Geriatr Soc. 2007; 55:824–831.
Article
37. Fernandes BS, Hodge JM, Pasco JA, et al. Effects of depression and serotonergic antidepressants on bone: mechanisms and implications for the treatment of depression. Drugs Aging. 2016; 33:21–25.
Article
38. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284:143–147.
Article
39. Vidal C, Li W, Santner-Nanan B, et al. The kynurenine pathway of tryptophan degradation is activated during osteoblastogenesis. Stem Cells. 2015; 33:111–121.
Article
40. Pälvimäki EP, Roth BL, Majasuo H, et al. Interactions of selective serotonin reuptake inhibitors with the serotonin 5-HT2c receptor. Psychopharmacology (Berl). 1996; 126:234–240.
Article
41. Kruk JS, Vasefi MS, Gondora N, et al. Fluoxetine-induced transactivation of the platelet-derived growth factor type beta receptor reveals a novel heterologous desensitization process. Mol Cell Neurosci. 2015; 65:45–51.
Article
42. Nam SS, Lee JC, Kim HJ, et al. Serotonin inhibits osteoblast differentiation and bone regeneration in rats. J Periodontol. 2016; 87:461–469.
Article
43. Bradaschia-Correa V, Josephson AM, Mehta D, et al. The selective serotonin reuptake inhibitor fluoxetine directly inhibits osteoblast differentiation and mineralization during fracture healing in mice. J Bone Miner Res. 2017; 32:821–833.
Article
Full Text Links
  • JBM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr