Skip to main content
Log in

Development of a Novel Perfusion Rotating Wall Vessel Bioreactor with Ultrasound Stimulation for Mass-Production of Mineralized Tissue Constructs

  • Original Article
  • Published:
Tissue Engineering and Regenerative Medicine Aims and scope

Abstract

Background:

As stem cells are considered a promising cell source for tissue engineering, many culture strategies have been extensively studied to generate in vitro stem cell-based tissue constructs. However, most approaches using conventional tissue culture plates are limited by the lack of biological relevance in stem cell microenvironments required for neotissue formation. In this study, a novel perfusion rotating wall vessel (RWV) bioreactor was developed for mass-production of stem cell-based 3D tissue constructs.

Methods:

An automated RWV bioreactor was fabricated, which is capable of controlling continuous medium perfusion, highly efficient gas exchange with surrounding air, as well as low-intensity pulsed ultrasound (LIPUS) stimulation. Embryonic stem cells encapsulated in alginate/gelatin hydrogel were cultured in the osteogenic medium by using our bioreactor system. Cellular viability, growth kinetics, and osteogenesis/mineralization were thoroughly evaluated, and culture media were profiled at real time. The in vivo efficacy was examined by a rabbit cranial defect model.

Results:

Our bioreactor successfully maintained the optimal culture environments for stem cell proliferation, osteogenic differentiation, and mineralized tissue formation during the culture period. The mineralized tissue constructs produced by our bioreactor demonstrated higher void filling efficacy in the large bone defects compared to the group implanted with hydrogel beads only. In addition, the LIPUS modules mounted on our bioreactor successfully reached higher mineralization of the tissue constructs compared to the groups without LIPUS stimulation.

Conclusion:

This study suggests an effective biomanufacturing strategy for mass-production of implantable mineralized tissue constructs from stem cells that could be applicable to future clinical practice.

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
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Polak JM, Mantalaris S. Stem cells bioprocessing: An important milestone to move regenerative medicine research into the clinical arena. Pediatr Res. 2008;63:461–6.

    Article  PubMed  Google Scholar 

  2. Peroglio M, Gaspar D, Zeugolis DI, Alini M. Relevance of bioreactors and whole tissue cultures for the translation of new therapies to humans. J Orthop Res. 2018;36:10–21.

    PubMed  Google Scholar 

  3. Cha JM, Shin EK, Sung JH, Moon GJ, Kim EH, Cho YH, et al. Efficient scalable production of therapeutic microvesicles derived from human mesenchymal stem cells. Sci Rep. 2018;8:1171.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Gieseck RL 3rd, Hannan NR, Bort R, Hanley NA, Drake RA, Cameron GW, et al. Maturation of induced pluripotent stem cell derived hepatocytes by 3D-culture. PloS One. 2014;9:e86372.

    Article  PubMed Central  CAS  Google Scholar 

  5. Xie Y, Hardouin P, Zhu Z, Tang T, Dai K, Lu J. Three-dimensional flow perfusion culture system for stem cell proliferation inside the critical-size beta-tricalcium phosphate scaffold. Tissue Eng. 2006;12:3535–43.

    Article  CAS  PubMed  Google Scholar 

  6. Zhao F, Pathi P, Grayson W, Xing Q, Locke BR, Ma T. Effects of oxygen transport on 3-d human mesenchymal stem cell metabolic activity in perfusion and static cultures: experiments and mathematical model. Biotechnol Prog. 2005;21:1269–80.

    Article  CAS  PubMed  Google Scholar 

  7. Cha JM, Mantalaris A, Jung S, Ji Y, Bang OY, Bae H. Mesoderm lineage 3D tissue constructs are produced at large-scale in a 3D stem cell bioprocess. Biotechnol J. 2017;12:1600748.

    Article  CAS  Google Scholar 

  8. Ishaug SL, Crane GM, Miller MJ, Yasko AW, Yaszemski MJ, Mikos AG. Bone formation by three-dimensional stromal osteoblast culture in biodegradable polymer scaffolds. J Biomed Mater Res. 1997;36:17–28.

    Article  CAS  Google Scholar 

  9. Martin I, Obradovic B, Freed LE, Vunjak-Novakovic G. Method for quantitative analysis of glycosaminoglycan distribution in cultured natural and engineered cartilage. Ann Biomed Eng. 1999;27:656–62.

    Article  CAS  PubMed  Google Scholar 

  10. Cameron CM, Hu WS, Kaufman DS. Improved development of human embryonic stem cell-derived embryoid bodies by stirred vessel cultivation. Biotechnol Bioeng. 2006;94:938–48.

    Article  CAS  PubMed  Google Scholar 

  11. Fok EY, Zandstra PW. Shear-controlled single-step mouse embryonic stem cell expansion and embryoid body-based differentiation. Stem Cells. 2005;23:1333–42.

    Article  CAS  PubMed  Google Scholar 

  12. Kehoe DE, Lock LT, Parikh A, Tzanakakis ES. Propagation of embryonic stem cells in stirred suspension without serum. Biotechnol Prog. 2008;24:1342–52.

    Article  CAS  PubMed  Google Scholar 

  13. Niebruegge S, Bauwens CL, Peerani R, Thavandiran N, Masse S, Sevaptisidis E, et al. Generation of human embryonic stem cell-derived mesoderm and cardiac cells using size-specified aggregates in an oxygen-controlled bioreactor. Biotechnol Bioeng. 2009;102:493–507.

    Article  CAS  Google Scholar 

  14. Niebruegge S, Nehring A, Bär H, Schroeder M, Zweigerdt R, Lehmann J. Cardiomyocyte production in mass suspension culture: embryonic stem cells as a source for great amounts of functional cardiomyocytes. Tissue Eng Part A. 2008;14:1591–601.

    Article  CAS  PubMed  Google Scholar 

  15. Schroeder M, Niebruegge S, Werner A, Willbold E, Burg M, Ruediger M, et al. Differentiation and lineage selection of mouse embryonic stem cells in a stirred bench scale bioreactor with automated process control. Biotechnol Bioeng. 2005;92:920–33.

    Article  CAS  PubMed  Google Scholar 

  16. Abranches E, Bekman E, Henrique D, Cabral JM. Expansion of mouse embryonic stem cells on microcarriers. Biotechnol Bioeng. 2007;96:1211–21.

    Article  CAS  PubMed  Google Scholar 

  17. Dang SM, Gerecht-Nir S, Chen J, Itskovitz-Eldor J, Zandstra PW. Controlled, scalable embryonic stem cell differentiation culture. Stem Cells. 2004;22:275–82.

    Article  PubMed  Google Scholar 

  18. Wang X, Wei G, Yu W, Zhao Y, Yu X, Ma X. Scalable producing embryoid bodies by rotary cell culture system and constructing engineered cardiac tissue with ES-derived cardiomyocytes in vitro. Biotechnol Prog. 2006;22:811–8.

    Article  PubMed  CAS  Google Scholar 

  19. Placzek MR, Chung IM, Macedo HM, Ismail S, Mortera Blanco T, Lim M, et al. Stem cell bioprocessing: fundamentals and principles. J R Soc Interface. 2009;6:209–32.

    Article  CAS  PubMed  Google Scholar 

  20. Dang SM, Kyba M, Perlingeiro R, Daley GQ, Zandstra PW. Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems. Biotechnol Bioeng. 2002;78:442–53.

    Article  CAS  PubMed  Google Scholar 

  21. Freed LE, Vunjaknovakovic G. Cultivation of cell-polymer tissue constructs in simulated microgravity. Biotechnol Bioeng. 1995;46:306–13.

    Article  CAS  PubMed  Google Scholar 

  22. Gerecht-Nir S, Cohen S, Itskovitz-Eldor J. Bioreactor cultivation enhances the efficiency of human embryoid body (hEB) formation and differentiation. Biotechnol Bioeng. 2004;86:493–502.

    Article  CAS  PubMed  Google Scholar 

  23. Hwang YS, Cho J, Tay F, Heng JY, Ho R, Kazarian SG, et al. The use of murine embryonic stem cells, alginate encapsulation, and rotary microgravity bioreactor in bone tissue engineering. Biomaterials. 2009;30:499–507.

    Article  CAS  PubMed  Google Scholar 

  24. Lü S, Liu S, He W, Duan C, Li Y, Liu Z, et al. Bioreactor cultivation enhances NTEB formation and differentiation of NTES cells into cardiomyocytes. Cloning Stem Cells. 2008;10:363–70.

    Article  PubMed  Google Scholar 

  25. Randle WL, Cha JM, Hwang YS, Chan KL, Kazarian SG, Polak JM, et al. Integrated 3-dimensional expansion and osteogenic differentiation of murine embryonic stem cells. Tissue Eng. 2007;13:2957–70.

    Article  CAS  PubMed  Google Scholar 

  26. Siti-Ismail N, Bishop AE, Polak JM, Mantalaris A. The benefit of human embryonic stem cell encapsulation for prolonged feeder-free maintenance. Biomaterials. 2008;29:3946–52.

    Article  CAS  PubMed  Google Scholar 

  27. Liu M, Liu N, Zang R, Li Y, Yang ST. Engineering stem cell niches in bioreactors. World J Stem Cells. 2013;5:124–35.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cha JM, Lee MY, Hong J. Bioreactor systems are essentially required for stem cell bioprocessing. Precis Future Med. 2019;3:19–23.

    Article  CAS  Google Scholar 

  29. de Peppo GM, Marcos-Campos I, Kahler DJ, Alsalman D, Shang L, Vunjak-Novakovic G, et al. Engineering bone tissue substitutes from human induced pluripotent stem cells. Proc Natl Acad Sci U S A. 2013;110:8680–5.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Marolt D, Campos IM, Bhumiratana S, Koren A, Petridis P, Zhang G, et al. Engineering bone tissue from human embryonic stem cells. Proc Natl Acad Sci U S A. 2012;109:8705–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Panciera T, Azzolin L, Cordenonsi M, Piccolo S. Mechanobiology of YAP and TAZ in physiology and disease. Nat Rev Mol Cell Biol. 2017;18:758–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yavropoulou MP, Yovos JG. The molecular basis of bone mechanotransduction. J Musculoskelet Neuronal Interact. 2016;16:221–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Perry MJ, Parry LK, Burton VJ, Gheduzzi S, Beresford JN, Humphrey VF, et al. Ultrasound mimics the effect of mechanical loading on bone formation in vivo on rat ulnae. Med Eng Phys. 2009;31:42–7.

    Article  PubMed  Google Scholar 

  34. Padilla F, Puts R, Vico L, Raum K. Stimulation of bone repair with ultrasound: a review of the possible mechanic effects. Ultrasonics. 2014;54:1125–45.

    Article  CAS  PubMed  Google Scholar 

  35. Rutten S, Nolte PA, Guit GL, Bouman DE, Albers GH. Use of low-intensity pulsed ultrasound for posttraumatic nonunions of the tibia: a review of patients treated in the Netherlands. J Trauma. 2007;62:902–8.

    PubMed  Google Scholar 

  36. Gilbert SJ, Stevens J, Vishwakarma AR, Archer CW, Duance VC. The use of pulsed low-intensity ultrasound (PLIUS) as a therapy for cartilage repair. Int J Exp Pathol. 2014;95:A12.

    Google Scholar 

  37. Calik M, Calik SG, Yilmaz H, Karaca G, Esen H, Avunduk MC, et al. Using low-intensity pulsed ultrasound to stimulate the consolidation of rib fractures: an experimental study. Eur Clin Respir J. 2014;44:1924.

    Google Scholar 

  38. Harrison A, Lin S, Pounder N, Mikuni-Takagaki Y. Mode & mechanism of low intensity pulsed ultrasound (LIPUS) in fracture repair. Ultrasonics. 2016;70:45–52.

    Article  CAS  PubMed  Google Scholar 

  39. Hwang YS, Randle WL, Bielby RC, Polak JM, Mantalaris A. Enhanced derivation of osteogenic cells from murine embryonic stem cells after treatment with HepG2-conditioned medium and modulation of the embryoid body formation period: application to skeletal tissue engineering. Tissue Eng. 2006;12:1381–92.

    Article  CAS  PubMed  Google Scholar 

  40. Rathjen J, Lake JA, Bettess MD, Washington JM, Chapman G, Rathjen PD. Formation of a primitive ectoderm like cell population, EPL cells, from ES cells in response to biologically derived factors. J Cell Sci. 1999;112:601–12.

    Article  CAS  PubMed  Google Scholar 

  41. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–8.

    Article  CAS  PubMed  Google Scholar 

  42. Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction. Anal Biochem. 2004;329:77–84.

    Article  CAS  PubMed  Google Scholar 

  43. Kazarian SG, Chan KLA. “Chemical photography” of drug release. Macromolecules. 2003;36:9866–72.

    Article  CAS  Google Scholar 

  44. Zhang GJ, Moore DJ, Flach CR, Mendelsohn R. Vibrational microscopy and imaging of skin: from single cells to intact tissue. Anal Bioanal Chem. 2007;387:1591–9.

    Article  CAS  PubMed  Google Scholar 

  45. Andrei DC, Briscoe BJ, Luckham PF, Williams DR. The deformation of microscopic gel particles. J Chim Phys Physicochim Biol. 1996;93:960–76.

    Article  CAS  Google Scholar 

  46. Briscoe BJ, Liu KK, Williams DR. Adhesive contact deformation of a single microelastomeric sphere. J Colloid Interface Sci. 1998;200:256–64.

    Article  CAS  Google Scholar 

  47. Karp JM, Ferreira LS, Khademhosseini A, Kwon AH, Yeh J, Langer RS. Cultivation of human embryonic stem cells without the embryoid body step enhances osteogenesis in vitro. Stem Cells. 2006;24:835–43.

    Article  PubMed  Google Scholar 

  48. Bauwens C, Yin T, Dang S, Peerani R, Zandstra PW. Development of a perfusion fed bioreactor for embryonic stem cell-derived cardiomyocyte generation: oxygen-mediated enhancement of cardiomyocyte output. Biotechnol Bioeng. 2005;90:452–61.

    Article  CAS  PubMed  Google Scholar 

  49. Cormier JT, Zur Nieden NI, Rancourt DE, Kallos MS. Expansion of undifferentiated murine embryonic stem cells as aggregates in suspension culture bioreactors. Tissue Eng. 2006;12:3233–45.

    Article  CAS  PubMed  Google Scholar 

  50. Smith AG. Mouse embryo stem cells: their identification, propagation and manipulation. Semin Cell Biol. 1992;3:385–99.

    Article  CAS  PubMed  Google Scholar 

  51. Conaghan J, Handyside AH, Winston RM, Leese HJ. Effects of pyruvate and glucose on the development of human preimplantation embryos in vitro. Reprod Fertil. 1993;99:87–95.

    Article  CAS  Google Scholar 

  52. Quinn P. Enhanced results in mouse and human embryo culture using a modified human tubal fluid medium lacking glucose and phosphate. J Assist Reprod Genet. 1995;12:97–105.

    Article  CAS  PubMed  Google Scholar 

  53. Khoo ML, McQuade LR, Smith MSR, Lees JG, Sidhu KS, Tuch BE. Growth and differentiation of embryoid bodies derived from human embryonic stem cells: effect of glucose and basic fibroblast growth factor. Biol Reprod. 2005;73:1147–56.

    Article  CAS  PubMed  Google Scholar 

  54. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell. 2002;108:17–29.

    Article  CAS  PubMed  Google Scholar 

  55. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation. Cell. 1997;89:747–54.

    Article  CAS  PubMed  Google Scholar 

  56. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell. 1997;89:755–64.

    Article  CAS  PubMed  Google Scholar 

  57. Zhang S, Cheng J, Qin YX. Mechanobiological modulation of cytoskeleton and calcium influx in osteoblastic cells by short-term focused acoustic radiation force. PLoS One. 2012;7:e38343.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by Incheon National University (International Cooperative) Research Grant in 2019.

Author information

Authors and Affiliations

Authors

Contributions

Jae Min Cha, PhD and Athanasios Mantalaris, PhD: Administrative support, Conception and design, Manuscript writing, review & editing. Yu-Shik Hwang, PhD, Dong-Ku Kang, PhD, and Elana S Cooper, MSc: Manuscript writing, Data analysis and interpretation. Jun Lee, DDS, PhD: Collection and assembly of data.

Corresponding authors

Correspondence to Jae Min Cha or Athanasios Mantalaris.

Ethics declarations

Conflict of interest

No competing financial interests exist.

Ethical statement

The animal studies were performed after receiving approval of the Institutional Animal Care and Use Committee (IACUC) in Wonkwang University (IACUC approval No. WKU21-37).

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

13770_2022_447_MOESM1_ESM.tif

Supplementary Figure S1. Surgery procedures and biopsies of cranial vault at 1, 2, and 8 weeks after operation. After the central region was incised (A), four holes (diameter: 8 mm, depth: 2-3 mm) were generated as the standardized cranial defects (B). The defects were filled with osteogenic constructs generated from the BSEL bioreactor and alginate beads (about 10 beads each), respectively (C). Finally, the defects were covered with fibrin glue in order to fix all the grafts within the holes (D). The defected cranial vaults were isolated at 1, 2, and 8 weeks after the implant insertion, and photographed to visually examine the inflammation around the defected sites (E). (TIF 1454 KB)

13770_2022_447_MOESM2_ESM.tif

Supplementary Figure S2. (A-C) Customized-design of the BSEL bioreactor for mounting the LIPUS modules consisting of four ultrasound probes that were evenly placed to the front part of the culture chamber to ensure uniform transmission of ultrasound. The ultrasound probes were inserted to the culture vessel to ensure the maximum ultrasound transmitting level and attached on a bearing connected to the culture vessel to rotate simultaneously. In order to avoid the effect of ultrasound reflection, the back of the culture chamber was fitted with the ultrasound absorber material, HAM-A. (D) The controller is an analog box whereby the LIPUS intensity, PRF, duty ratio, and time can be adjusted. (E and F) The effective area was obtained from the number of pixels and the effective radius was calculated from the circle area equation, representing the ultrasound uniformity until reaching 20 mm in depth and then expanded without significant change of ultrasound intensity. (TIF 651 KB)

13770_2022_447_MOESM3_ESM.tif

Supplementary Figure S3. Results of a pilot animal testing with mineralized beads produced by the BSEL bioreactor. About ten mineralized beads were implanted in each cranial defect (diameter 8 mm × depth 2-3 mm). As a control, alginate beads without cells were implanted. At eight weeks, all implanted tissues were biopsied for 3D micro-CT scanning, H&E staining, and immunohistochemistry for osteocalcin. (A and B) 3D-reconstructed micro-CT scanning data of the bone defects implanted with mineralized beads and alginate beads, respectively. (C and D) Histological (I: H&E staining) and immunohistochemical (II: osteocalcin) data of the bone defects implanted with mineralized beads and alginate beads, respectively. (NB: new bone, FI: fibrous interzone, DM: dura mater, arrows: traces of vascularization). Scale bars represent 500 µm (×50) in H&E staining and 250 µm (×100) in immunohistochemical data. (TIF 1338 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cha, J.M., Hwang, YS., Kang, DK. et al. Development of a Novel Perfusion Rotating Wall Vessel Bioreactor with Ultrasound Stimulation for Mass-Production of Mineralized Tissue Constructs. Tissue Eng Regen Med 19, 739–754 (2022). https://doi.org/10.1007/s13770-022-00447-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13770-022-00447-3

Keywords

Navigation