Skip to main content
Log in

Proof of Concept on Functionality Improvement of Mesenchymal Stem-Cells, in Postmenopausal Osteoporotic Women Treated with Teriparatide (PTH1-34), After Suffering Atypical Fractures

  • Original Research
  • Published:
Calcified Tissue International Aims and scope Submit manuscript

Abstract

Osteoporosis long-term treatment with nitrogen-containing bisphosphonates, has been associated with uncommon adverse effects, as atypical femoral fractures (AFF). Thus, treatment with teriparatide (TPTD; fragment of human parathyroid hormone; PTH1−34) has been proposed for such patients. Besides its anabolizing effect on bone, TPTD may affect stem-cell mobilization and expansion. Bone marrow mononuclear cells (BMMNC) were isolated from five women that had suffered AFF associated to bisphosphonate treatment, before and after 6 months of TPTD therapy. The presence of mesenchymal stromal cells (CD73, CD90 and CD105 positive cells), gene expression of NANOG, SOX2 and OCT4, proliferation, senescence and capacity to differentiate into osteoblasts and adipocytes were analyzed. After TPTD treatment, BMMNC positive cells for CD73, CD90 and CD105 increased from 6.5 to 37.5% (p < 0.05); NANOG, SOX2 and OCT4 were upregulated, being statistically significant for NANOG (p < 0.05), and cells increased proliferative capacity more than 50% at day 7 (p < 0.05). Senescence was reduced 2.5-fold (p < 0.05), increasing differentiation capacity into osteoblasts and adipocytes, with more than twice mineralization capacity of extracellular matrix or fat-droplet formation (p < 0.05), respectively. Results show that TPTD treatment caused BMMNC “rejuvenation”, increasing the number of cells in a more undifferentiated stage, with higher differentiation potency. This effect may favor TPTD anabolic action on bone in such patients with AFF, increasing osteoblast precursor cells. Such response could also arise in other osteoporotic patients treated with TPTD, without previous AFF. Furthermore, our data suggest that TPTD effect on stromal cells may have clinical implications for bone-regenerative medicine. Further studies may deepen on this potential.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Klibanski A, Adams-Campbell L, Bassford T et al (2001) Osteoporosis prevention, diagnosis, and therapy. JAMA 285:785–795

    Article  Google Scholar 

  2. Cauley JA (2013) Public health impact of osteoporosis. J Gerontol Ser A 68:1243–1251. https://doi.org/10.1093/gerona/glt093

    Article  Google Scholar 

  3. Diab DL, Watts NB (2012) Bisphosphonates in the treatment of osteoporosis. Endocrinol Metab Clin North Am 41:487–506. https://doi.org/10.1016/j.ecl.2012.04.007

    Article  CAS  PubMed  Google Scholar 

  4. Freemantle N, Cooper C, Diez-Perez A et al (2013) Results of indirect and mixed treatment comparison of fracture efficacy for osteoporosis treatments: a meta-analysis. Osteoporos Int 24:209–217. https://doi.org/10.1007/s00198-012-2068-9

    Article  CAS  PubMed  Google Scholar 

  5. Kennel KA, Drake MT (2009) Adverse effects of bisphosphonates: implications for osteoporosis management. Mayo Clin Proc 84:632–638. https://doi.org/10.1016/S0025-6196(11)60752-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shane E, Burr D, Abrahamsen B et al (2014) Atypical subtrochanteric and diaphyseal femoral fractures: second report of a task force of the American society for bone and mineral research. J Bone Miner Res 29:1–23. https://doi.org/10.1002/jbmr.1998

    Article  PubMed  Google Scholar 

  7. Roca-Ayats N, Balcells S, Garcia-Giralt N et al (2017) GGPS1 mutation and atypical femoral fractures with bisphosphonates. N Engl J Med 376:1794–1795. https://doi.org/10.1056/NEJMc1612804

    Article  PubMed  Google Scholar 

  8. Nguyen HH, Milat F, Ebeling PR (2017) Bone Reports Case Report A new contralateral atypical femoral fracture despite sequential therapy with teriparatide and strontium ranelate. Bone Rep 6:34–37. https://doi.org/10.1016/j.bonr.2017.01.002

    Article  PubMed  PubMed Central  Google Scholar 

  9. Pietrogrande L, Raimondo E (2013) Teriparatide in the treatment of non-unions: scientific and clinical evidences. Injury 44(Suppl 1):S54–S57. https://doi.org/10.1016/S0020-1383(13)70013-5

    Article  PubMed  Google Scholar 

  10. Mastaglia SR, Aguilar G, Oliveri B (2016) Teriparatide for the rapid resolution of delayed healing of atypical fractures associated with long-term bisphosphonate use. Eur J Rheumatol 3:87–90. https://doi.org/10.5152/eurjrheum.2015.0010

    Article  PubMed  Google Scholar 

  11. Jilka RL (2007) Molecular and cellular mechanisms of the anabolic effect of intermittent PTH. Bone 40:1434–1446. https://doi.org/10.1016/j.bone.2007.03.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kim SW, Pajevic PD, Selig M et al (2012) Intermittent parathyroid hormone administration converts quiescent lining cells to active osteoblasts. J Bone Miner Res 27:2075–2084. https://doi.org/10.1002/jbmr.1665

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Casado-Díaz A, Quesada-Gómez JM, Dorado G (2016) Stem cell research and molecular markers in medicine. In: Caplan M (ed) Reference module in biomedical sciences. Biochemistry, cell biology and molecular biology. Elsevier, Amsterdam, p 14. https://doi.org/10.1016/B978-0-12-801238-3

    Chapter  Google Scholar 

  14. Zaim M, Karaman S, Cetin G, Isik S (2012) Donor age and long-term culture affect differentiation and proliferation of human bone marrow mesenchymal stem cells. Ann Hematol 91:1175–1186. https://doi.org/10.1007/s00277-012-1438-x

    Article  PubMed  Google Scholar 

  15. Moerman EJ, Teng K, Lipschitz D, Lecka-Czernik B (2004) Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGF-beta/BMP signaling pathways. Aging Cell 3:379–389. https://doi.org/10.1111/j.1474-9728.2004.00127.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Singh L, Brennan TA, Russell E et al (2016) Aging alters bone-fat reciprocity by shifting in vivo mesenchymal precursor cell fate towards an adipogenic lineage. Bone 85:29–36. https://doi.org/10.1016/j.bone.2016.01.014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Casado-Díaz A, Santiago-Mora R, Jiménez R et al (2008) Cryopreserved human bone marrow mononuclear cells as a source of mesenchymal stromal cells: application in osteoporosis research. Cytotherapy 10:460–468. https://doi.org/10.1080/14653240802192644

    Article  CAS  PubMed  Google Scholar 

  18. D’Amelio P, Tamone C, Sassi F et al (2012) Teriparatide increases the maturation of circulating osteoblast precursors. Osteoporos Int 23:1245–1253. https://doi.org/10.1007/s00198-011-1666-2

    Article  CAS  PubMed  Google Scholar 

  19. Huber BC (2014) Impact of parathyroid hormone on bone marrow-derived stem cell mobilization and migration. World J Stem Cells 6:637. https://doi.org/10.4252/wjsc.v6.i5.637

    Article  PubMed  PubMed Central  Google Scholar 

  20. Di Bernardo G, Galderisi U, Fiorito C et al (2010) Dual role of parathyroid hormone in endothelial progenitor cells and marrow stromal mesenchymal stem cells. J Cell Physiol 222:474–480. https://doi.org/10.1002/jcp.21976

    Article  CAS  PubMed  Google Scholar 

  21. Feron J-M, Cambon-Binder A (2017) Medication management after intramedullary nailing of atypical fractures. Injury 48 Suppl 1:S15–S17. https://doi.org/10.1016/j.injury.2017.04.030

  22. Vasikaran S, Eastell R, Bruyère O et al (2011) Markers of bone turnover for the prediction of fracture risk and monitoring of osteoporosis treatment: a need for international reference standards. Osteoporos Int 22:391–420. https://doi.org/10.1007/s00198-010-1501-1

    Article  CAS  PubMed  Google Scholar 

  23. Güerri-Fernández RC, Nogués X, Quesada Gómez JM et al (2013) Microindentation for in vivo measurement of bone tissue material properties in atypical femoral fracture patients and controls. J Bone Miner Res 28:162–168. https://doi.org/10.1002/jbmr.1731

    Article  CAS  PubMed  Google Scholar 

  24. Quesada-Gomez JM, Bouillon R (2018) Is calcifediol better than cholecalciferol for vitamin D supplementation? Osteoporos Int 29:1697–1711. https://doi.org/10.1007/s00198-018-4520-y

    Article  CAS  PubMed  Google Scholar 

  25. Gregory CA, Gunn WG, Peister A, Prockop DJ (2004) An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction. Anal Biochem 329:77–84. https://doi.org/10.1016/j.ab.2004.02.002

    Article  CAS  PubMed  Google Scholar 

  26. Dominici M, Le Blanc K, Mueller I et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317. https://doi.org/10.1080/14653240600855905

    Article  CAS  Google Scholar 

  27. Calloni R, Cordero EAA, Henriques JAP, Bonatto D (2013) Reviewing and updating the major molecular markers for stem cells. Stem Cells Dev 22:1455–1476. https://doi.org/10.1089/scd.2012.0637

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Das S, Levasseur D (2013) Transcriptional regulatory mechanisms that govern embryonic stem cell fate. Methods Mol Biol 1029:191–203. https://doi.org/10.1007/978-1-62703-478-4_13

    Article  CAS  PubMed  Google Scholar 

  29. Bais MV, Shabin ZM, Young M et al (2012) Role of Nanog in the maintenance of marrow stromal stem cells during post natal bone regeneration. Biochem Biophys Res Commun 417:211–216. https://doi.org/10.1016/j.bbrc.2011.11.087

    Article  CAS  PubMed  Google Scholar 

  30. Han J, Mistriotis P, Lei P et al (2012) Nanog reverses the effects of organismal aging on mesenchymal stem cell proliferation and myogenic differentiation potential. Stem Cells 30:2746–2759. https://doi.org/10.1002/stem.1223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Han S-M, Han S-H, Coh Y-R et al (2014) Enhanced proliferation and differentiation of Oct4- and Sox2-overexpressing human adipose tissue mesenchymal stem cells. Exp Mol Med 46:e101. https://doi.org/10.1038/emm.2014.28

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yoon DS, Kim YH, Jung HS et al (2011) Importance of Sox2 in maintenance of cell proliferation and multipotency of mesenchymal stem cells in low-density culture. Cell Prolif 44:428–440. https://doi.org/10.1111/j.1365-2184.2011.00770.x

    Article  CAS  PubMed  Google Scholar 

  33. Guo L, Qi S-T, Miao D-Q et al (2012) The roles of parathyroid hormone-like hormone during mouse preimplantation embryonic development. PLoS ONE 7:e40528. https://doi.org/10.1371/journal.pone.0040528

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jüppner H, Abou-Samra AB, Freeman M et al (1991) A G protein-linked receptor for parathyroid hormone and parathyroid hormone-related peptide. Science 254:1024–1026

    Article  PubMed  Google Scholar 

  35. Zhou S, Greenberger JS, Epperly MW et al (2008) Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell 7:335–343. https://doi.org/10.1111/j.1474-9726.2008.00377.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bellantuono I, Aldahmash A, Kassem M (2009) Aging of marrow stromal (skeletal) stem cells and their contribution to age-related bone loss. Biochim Biophys Acta 1792:364–370. https://doi.org/10.1016/j.bbadis.2009.01.008

    Article  CAS  PubMed  Google Scholar 

  37. Stenderup K, Justesen J, Clausen C, Kassem M (2003) Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone 33:919–926

    Article  Google Scholar 

  38. Fickert S, Schröter-Bobsin U, Gross A-F et al (2011) Human mesenchymal stem cell proliferation and osteogenic differentiation during long-term ex vivo cultivation is not age dependent. J Bone Miner Metab 29:224–235. https://doi.org/10.1007/s00774-010-0215-y

    Article  PubMed  Google Scholar 

  39. Chen Q, Shou P, Zheng C et al (2016) Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. https://doi.org/10.1038/cdd.2015.168

    Article  PubMed  PubMed Central  Google Scholar 

  40. Rickard DJ, Wang F-L, Rodriguez-Rojas A-M et al (2006) Intermittent treatment with parathyroid hormone (PTH) as well as a non-peptide small molecule agonist of the PTH1 receptor inhibits adipocyte differentiation in human bone marrow stromal cells. Bone 39:1361–1372. https://doi.org/10.1016/j.bone.2006.06.010

    Article  CAS  PubMed  Google Scholar 

  41. Chandra A, Lan S, Zhu J et al (2013) PTH prevents the adverse effects of focal radiation on bone architecture in young rats. Bone 55:449–457. https://doi.org/10.1016/j.bone.2013.02.023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Geng S, Zhou S, Glowacki J (2011) Age-related decline in osteoblastogenesis and 1α-hydroxylase/CYP27B1 in human mesenchymal stem cells: stimulation by parathyroid hormone. Aging Cell 10:962–971. https://doi.org/10.1111/j.1474-9726.2011.00735.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Garrett RW, Emerson SG (2008) The role of parathyroid hormone and insulin-like growth factors in hematopoietic niches: physiology and pharmacology. Mol Cell Endocrinol 288:6–10. https://doi.org/10.1016/j.mce.2008.02.022

    Article  CAS  PubMed  Google Scholar 

  44. Yao H, Miura Y, Yoshioka S et al (2014) Parathyroid hormone enhances hematopoietic expansion via upregulation of cadherin-11 in bone marrow mesenchymal stromal cells. Stem Cells 32:2245–2255. https://doi.org/10.1002/stem.1701

    Article  CAS  PubMed  Google Scholar 

  45. Wang L-L, Chen D, Lee J et al (2014) Mobilization of endogenous bone marrow derived endothelial progenitor cells and therapeutic potential of parathyroid hormone after ischemic stroke in mice. PLoS ONE 9:e87284. https://doi.org/10.1371/journal.pone.0087284

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Langdahl BL, Ljunggren Ö, Benhamou CL et al (2016) Fracture rate, quality of life and back pain in patients with osteoporosis treated with teriparatide: 24-month results from the extended forsteo observational study (ExFOS). Calcif Tissue Int 99:259–271. https://doi.org/10.1007/s00223-016-0143-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge all of the funding sources.

Funding

Supported by Grants AGL2013-45110-R of “Ministerio de Ciencia e Innovación” (MICINN); PI081692 and PI15/01857 “Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad” (RETICEF) and CIBER “Fragilidad y Envejecimiento Saludable” (CIBERFES) of “Instituto de Salud Carlos III” (ISCIII), “Ministerio de Economía y Competitividad” (MINECO) and European Union (EU); and “Ayudas de Intensificación de la Investigación” and “Grupo CTS413” of “Junta de Andalucía”, Spain.

Author information

Authors and Affiliations

Authors

Contributions

ACD, GD and JMQG conceived and designed the experiments. ACD, MG, MJM and CNV performed the experiments. ACD, ADP and JMQG contributed with analysis tools and patients data. ACD, GD and JMQG wrote the paper. All authors analyzed and interpreted the data. JMQG is guarantor.

Corresponding author

Correspondence to José Manuel Quesada-Gómez.

Ethics declarations

Conflict of interest

Antonio Casado-Díaz, Gabriel Dorado, Mercè Giner, María José Montoya, Cristina Navarro-Valverde, Adolfo Díez-Pérez, José Manuel Quesada-Gómez declare no conflict of interest.

Ethics Approval

Patients signed informed consent, in accordance with regulations of the Clinical Research Ethics Committee of Parc de Salut Mar, which approved the study. This was carried out in accordance with the terms of the Declaration of Helsinki.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Casado-Díaz, A., Dorado, G., Giner, M. et al. Proof of Concept on Functionality Improvement of Mesenchymal Stem-Cells, in Postmenopausal Osteoporotic Women Treated with Teriparatide (PTH1-34), After Suffering Atypical Fractures. Calcif Tissue Int 104, 631–640 (2019). https://doi.org/10.1007/s00223-019-00533-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00223-019-00533-0

Keywords

Navigation