Skip to main content
Log in

Biofabrication of size-controlled liver microtissues incorporated with ECM-derived microparticles to prolong hepatocyte function

  • Research Article
  • Published:
Bio-Design and Manufacturing Aims and scope Submit manuscript

Abstract

Multicellular microtissues of primary human hepatocytes (PHHs) co-cultured with other supporting cell types are a promising model for drug screening and toxicological studies. However, these liver microtissues (LMs) rapidly lose their functions during ex vivo culture. Here, in order to mimic the cellular and structural hepatic microenvironment, we co-cultured PHHs with human mesenchymal stromal cells (MSCs) and human umbilical vein endothelial cells (HUVECs) in the presence of cell-sized microparticles (MPs) derived from liver extracellular matrix (LEMPs). The microwell culture platform enabled biofabrication of size-controlled multicellular microtissues (PHH:HUVEC:MSC = 3:2:1) with efficient LEMP incorporation (about 70% at a 2:1 ratio of cells:MP). The biofabricated liver microtissues (BLMs) were cultured ex vivo for 14 days and compared to the cell-only LM in terms of gene and protein expression, functional activity, cytochrome P450 (CYP450) enzyme inducibility, and drug sensitivity. The results supported superior hepatic-related gene expression, functional activity, and polarity for PHH in BLM compared to LM. CYP450 enzyme inducibility and dose-responsive sensitivity to toxic drugs were significantly higher in the BLM group. In conclusion, microtissue engineering by incorporation of tissue-specific microparticles within a multicellular microtissue can offer some advantages for drug discovery studies and cell transplantation applications. In the near future, this approach could generate a scalable platform of several functional biofabricated microtissues representing different organs.

Graphic abstract

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

Similar content being viewed by others

References

  1. Fraczek J, Bolleyn J, Vanhaecke T et al (2013) Primary hepatocyte cultures for pharmaco-toxicological studies: at the busy crossroad of various anti-dedifferentiation strategies. Arch Toxicol 87:577–610. https://doi.org/10.1007/s00204-012-0983-3

    Article  Google Scholar 

  2. Vinken M, Vanhaecke T, Rogiers V (2012) Primary hepatocyte cultures as in vitro tools for toxicity testing: quo vadis? Toxicol In Vitro 26:541–544. https://doi.org/10.1016/j.tiv.2012.01.002

    Article  Google Scholar 

  3. Heydari Z, Najimi M, Mirzaei H et al (2020) Tissue engineering in liver regenerative medicine: insights into novel translational technologies. Cells 9:304. https://doi.org/10.3390/cells9020304

    Article  Google Scholar 

  4. Zahmatkesh E, Khoshdel-Rad N, Mirzaei H et al (2021) Evolution of organoid technology: lessons learnt in Co-Culture systems from developmental biology. Dev Biol. https://doi.org/10.1016/j.ydbio.2021.03.001

    Article  Google Scholar 

  5. Heydari Z, Vosough M (2017) New platforms for drug screening and toxicology: necessity or need? Mod Med Lab J 1:107–109. https://doi.org/10.30699/mmlj17.1.3.107

    Article  Google Scholar 

  6. Friedman SL, Roll FJ, Boyles J et al (1985) Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. PNAS 82:8681–8685. https://doi.org/10.1073/pnas.82.24.8681

    Article  Google Scholar 

  7. Godoy P, Hewitt NJ, Albrecht U et al (2013) Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 87:1315–1530. https://doi.org/10.1007/s00204-013-1078-5

    Article  Google Scholar 

  8. Vinken M, Papeleu P, Snykers S et al (2006) Involvement of cell junctions in hepatocyte culture functionality. Crit Rev Toxicol 36:299–318. https://doi.org/10.1080/10408440600599273

    Article  Google Scholar 

  9. Bell CC, Hendriks DF, Moro SM et al (2016) Characterization of primary human hepatocyte spheroids as a model system for drug-induced liver injury, liver function and disease. Sci Rep 6:25187. https://doi.org/10.1038/srep25187

    Article  Google Scholar 

  10. Khetani SR, Bhatia SN (2008) Microscale culture of human liver cells for drug development. Nat Biotechnol 26:120. https://doi.org/10.1038/nbt1361

    Article  Google Scholar 

  11. Swift B, Pfeifer ND, Brouwer KL (2010) Sandwich-cultured hepatocytes: an in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity. Drug Metab Rev 42:446–471. https://doi.org/10.3109/03602530903491881

    Article  Google Scholar 

  12. Vernetti LA, Senutovitch N, Boltz R et al (2016) A human liver microphysiology platform for investigating physiology, drug safety, and disease models. Exp Biol Med 241:101–114. https://doi.org/10.1177/1535370215592121

    Article  Google Scholar 

  13. Vildhede A, Mateus A, Kha EK et al (2016) Mechanistic modeling of pitavastatin disposition in sandwich-cultured human hepatocytes: a proteomics-informed bottom-up approach. Drug Metab Dispos 44:505–516. https://doi.org/10.1124/dmd.115.066746

    Article  Google Scholar 

  14. Baze A, Parmentier C, Hendriks DF et al (2018) Three-dimensional spheroid primary human hepatocytes in monoculture and coculture with nonparenchymal cells. Tissue Eng C Methods 24:534–545. https://doi.org/10.1089/ten.tec.2018.0134

    Article  Google Scholar 

  15. Salerno S, Campana C, Morelli S et al (2011) Human hepatocytes and endothelial cells in organotypic membrane systems. Biomaterials 32:8848–8859. https://doi.org/10.1016/j.biomaterials.2011.08.004

    Article  Google Scholar 

  16. Ramachandran SD, Schirmer K, Münst B et al (2015) In vitro generation of functional liver organoid-like structures using adult human cells. PLoS ONE 10:e0139345. https://doi.org/10.1371/journal.pone.0139345

    Article  Google Scholar 

  17. Asai A, Aihara E, Watson C et al (2017) Paracrine signals regulate human liver organoid maturation from induced pluripotent stem cells. Development 144:1056–1064. https://doi.org/10.1242/dev.142794

    Article  Google Scholar 

  18. Muncie JM, Weaver VM (2018) The physical and biochemical properties of the extracellular matrix regulate cell fate. Curr Top Dev Biol 130:1–37. https://doi.org/10.1016/bs.ctdb.2018.02.002

    Article  Google Scholar 

  19. Hansen KC, Kiemele L, Maller O et al (2009) An in-solution ultrasonication-assisted digestion method for improved extracellular matrix proteome coverage. Mol Cell Proteom 8:1648–1657. https://doi.org/10.1074/mcp.m900039-mcp200

    Article  Google Scholar 

  20. Baptista PM, Siddiqui MM, Lozier G et al (2011) The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology 53:604–617. https://doi.org/10.1002/hep.24067

    Article  Google Scholar 

  21. Skardal A, Smith L, Bharadwaj S et al (2012) Tissue specific synthetic ECM hydrogels for 3-D in vitro maintenance of hepatocyte function. Biomaterials 33:4565–4575. https://doi.org/10.1016/j.biomaterials.2012.03.034

    Article  Google Scholar 

  22. Nakamura S, Ijima H (2013) Solubilized matrix derived from decellularized liver as a growth factor-immobilizable scaffold for hepatocyte culture. J Biosci Bioeng 116:746–753. https://doi.org/10.1016/j.jbiosc.2013.05.031

    Article  Google Scholar 

  23. Saheli M, Sepantafar M, Pournasr B et al (2018) Three-dimensional liver-derived extracellular matrix hydrogel promotes liver organoids function. J Cell Biochem 119:4320–4333. https://doi.org/10.1002/jcb.26622

    Article  Google Scholar 

  24. Darakhshan S, Pour AB, Kowsari-Esfahan R et al (2020) Generation of scalable hepatic micro-tissues as a platform for toxicological studies. Tissue Eng Regen Med 17:459–475. https://doi.org/10.1007/s13770-020-00272-6

    Article  Google Scholar 

  25. Ruoß M, Vosough M, Königsrainer A et al (2020) Towards improved hepatocyte cultures: progress and limitations. Food Chem Toxicol 138:111188. https://doi.org/10.1016/j.fct.2020.111188

    Article  Google Scholar 

  26. Jaffe EA, Nachman RL, Becker CG et al (1973) Culture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J Clin Investig 52:2745–2756. https://doi.org/10.1172/jci107470

    Article  Google Scholar 

  27. Zabulica M, Srinivasan RC, Vosough M et al (2019) Guide to the assessment of mature liver gene expression in stem cell-derived hepatocytes. Stem Cells Dev 28:907–919. https://doi.org/10.1089/scd.2019.0064

    Article  Google Scholar 

  28. Vosough M, Omidinia E, Kadivar M et al (2013) Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev 22:2693–2705. https://doi.org/10.1089/scd.2013.0088

    Article  Google Scholar 

  29. Gramignoli R, Tahan V, Dorko K et al (2014) Rapid and sensitive assessment of human hepatocyte functions. Cell Transplant 23:1545–1556. https://doi.org/10.3727/096368914x680064

    Article  Google Scholar 

  30. Collins SD, Yuen G, Tu T et al (2019) In vitro models of the liver: disease modeling, drug discovery and clinical applications. Exon Publ 3:47–67

    Google Scholar 

  31. Schutte M, Fox B, Baradez MO et al (2011) Rat primary hepatocytes show enhanced performance and sensitivity to acetaminophen during three-dimensional culture on a polystyrene scaffold designed for routine use. ASSAY Drug Dev Technol 9:475–486. https://doi.org/10.1089/adt.2011.0371

    Article  Google Scholar 

  32. Schyschka L, Sánchez JM, Wang Z et al (2013) Hepatic 3D cultures but not 2D cultures preserve specific transporter activity for acetaminophen-induced hepatotoxicity. Arch Toxicol 87:1581–1593. https://doi.org/10.1007/s00204-013-1080-y

    Article  Google Scholar 

  33. Bachmann A, Moll M, Gottwald E et al (2015) 3D cultivation techniques for primary human hepatocytes. Microarrays 4:64–83. https://doi.org/10.3390/microarrays4010064

    Article  Google Scholar 

  34. Ruoß M, Häussling V, Schügner F et al (2018) A standardized collagen-based scaffold improves human hepatocyte shipment and allows metabolic studies over 10 days. Bioengineering 5:86. https://doi.org/10.3390/bioengineering5040086

    Article  Google Scholar 

  35. Ranucci CS, Kumar A, Batra SP et al (2000) Control of hepatocyte function on collagen foams: sizing matrix pores toward selective induction of 2-D and 3-D cellular morphogenesis. Biomaterials 21:783–793. https://doi.org/10.1016/s0142-9612(99)00238-0

    Article  Google Scholar 

  36. Jain E, Damania A, Shakya AK et al (2015) Fabrication of macroporous cryogels as potential hepatocyte carriers for bioartificial liver support. Colloids Surf B Biointerfaces 136:761–771. https://doi.org/10.1016/j.colsurfb.2015.10.012

    Article  Google Scholar 

  37. Kumari J, Kumar A (2017) Development of polymer based cryogel matrix for transportation and storage of mammalian cells. Sci Rep 7:41551. https://doi.org/10.1038/srep41551

    Article  Google Scholar 

  38. Yamada M, Hori A, Sugaya S et al (2015) Cell-sized condensed collagen microparticles for preparing microengineered composite spheroids of primary hepatocytes. Lab Chip 15:3941–3951. https://doi.org/10.1039/c5lc00785b

    Article  Google Scholar 

  39. Ajoudanian M, Enomoto K, Tokunaga Y et al (2019) Self-organization of hepatocyte morphogenesis depending on the size of collagen microbeads relative to hepatocytes. Biofabrication 11:035007. https://doi.org/10.1088/1758-5090/ab145d

    Article  Google Scholar 

  40. Heidariyan Z, Ghanian MH, Ashjari M et al (2018) Efficient and cost-effective generation of hepatocyte-like cells through microparticle-mediated delivery of growth factors in a 3D culture of human pluripotent stem cells. Biomaterials 159:174–188. https://doi.org/10.1016/j.biomaterials.2018.01.005

    Article  Google Scholar 

  41. O’Brien MP, Carnes ME, Page RL et al (2016) Designing biopolymer microthreads for tissue engineering and regenerative medicine. Curr Stem Cell Rep 2:147–157. https://doi.org/10.1007/s40778-016-0041-9

    Article  Google Scholar 

  42. Martinez-Hernandez A, Amenta PS (1993) The hepatic extracellular matrix. Virchows Arch A 423:77–84. https://doi.org/10.1007/BF01606580

    Article  Google Scholar 

  43. Bhatia S, Balis U, Yarmush M et al (1999) Effect of cell–cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J 13:1883–1900. https://doi.org/10.1096/fasebj.13.14.1883

    Article  Google Scholar 

  44. Takebe T, Sekine K, Enomura M et al (2013) Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499:481–484. https://doi.org/10.1038/nature12271

    Article  Google Scholar 

  45. Ravindran S, Roam JL, Nguyen PK et al (2011) Changes of chondrocyte expression profiles in human MSC aggregates in the presence of PEG microspheres and TGF-β3. Biomaterials 32:8436–8445. https://doi.org/10.1016/j.biomaterials.2011.07.056

    Article  Google Scholar 

  46. Sj K, Kim EM, Yamamoto M et al (2020) Engineering multi-cellular spheroids for tissue engineering and regenerative medicine. Adv Healthc Mater 9:2000608. https://doi.org/10.1002/adhm.202000608

    Article  Google Scholar 

  47. You J, Park SA, Shin DS et al (2013) Characterizing the effects of heparin gel stiffness on function of primary hepatocytes. Tissue Eng A 19:2655–2663. https://doi.org/10.1089/ten.tea.2012.0681

    Article  Google Scholar 

  48. Mederacke I, Wursthorn K, Kirschner J et al (2009) Food intake increases liver stiffness in patients with chronic or resolved hepatitis C virus infection. Liver Int 29:1500–1506. https://doi.org/10.1111/j.1478-3231.2009.02100.x

    Article  Google Scholar 

  49. Desai SS, Tung JC, Zhou VX et al (2016) Physiological ranges of matrix rigidity modulate primary mouse hepatocyte function in part through hepatocyte nuclear factor 4 alpha. Hepatology 64:261–275. https://doi.org/10.1002/hep.28450

    Article  Google Scholar 

  50. Chen AA, Khetani SR, Lee S et al (2009) Modulation of hepatocyte phenotype in vitro via chemomechanical tuning of polyelectrolyte multilayers. Biomaterials 30:1113–1120. https://doi.org/10.1016/j.biomaterials.2008.10.055

    Article  Google Scholar 

  51. Riss T, Trask OJ (2021) Factors to consider when interrogating 3D culture models with plate readers or automated microscopes. Vitro Cell Dev Biol Anim 57:238–256. https://doi.org/10.1007/s11626-020-00537-3

    Article  Google Scholar 

  52. Sirenko O, Mitlo T, Hesley J et al (2015) High-content assays for characterizing the viability and morphology of 3D cancer spheroid cultures. ASSAY Drug Dev Technol 13:402–414. https://doi.org/10.1089/adt.2015.655

    Article  Google Scholar 

  53. Xu J, Lamouille S, Derynck R (2009) TGF-β-induced epithelial to mesenchymal transition. Cell Res 19:156–172. https://doi.org/10.1038/cr.2009.5

    Article  Google Scholar 

  54. Godoy P, Hengstler JG, Ilkavets I et al (2009) Extracellular matrix modulates sensitivity of hepatocytes to fibroblastoid dedifferentiation and transforming growth factor β–induced apoptosis. Hepatology 49:2031–2043. https://doi.org/10.1002/hep.22880

    Article  Google Scholar 

  55. LeCluyse EL (2001) Human hepatocyte culture systems for the in vitro evaluation of cytochrome P450 expression and regulation. Eur J Pharm Sci 13:343–368. https://doi.org/10.1016/s0928-0987(01)00135-x

    Article  Google Scholar 

  56. Hewitt NJ, Gómez Lechón MJ, Houston JB et al (2007) Primary hepatocytes: current understanding of the regulation of metabolic enzymes and transporter proteins, and pharmaceutical practice for the use of hepatocytes in metabolism, enzyme induction, transporter, clearance, and hepatotoxicity studies. Drug Metab Rev 39:159–234. https://doi.org/10.1080/03602530601093489

    Article  Google Scholar 

  57. Messner S, Agarkova I, Moritz W et al (2013) Multi-cell type human liver microtissues for hepatotoxicity testing. Arch Toxicol 87:209–213. https://doi.org/10.1007/s00204-012-0968-2

    Article  Google Scholar 

  58. Kostadinova R, Boess F, Applegate D et al (2013) A long-term three dimensional liver co-culture system for improved prediction of clinically relevant drug-induced hepatotoxicity. Toxicol Appl Pharmacol 268:1–16. https://doi.org/10.1016/j.taap.2013.01.012

    Article  Google Scholar 

  59. Jiang J, Messner S, Kelm J et al (2019) Human 3D multicellular microtissues: an upgraded model for the in vitro mechanistic investigation of inflammation-associated drug toxicity. Toxicol Lett 312:34–44. https://doi.org/10.1016/j.toxlet.2019.05.004

    Article  Google Scholar 

Download references

Acknowledgements

This project was financially supported by Grants from Royan Institute (No. 96000165) to MV and HB; Bahar Tashkhis Teb Co. (Nos. BTT, 9702, and 9802), Iran National Science Foundation (No. 97014445) to MV; the Ministry of Health and Medical Education (No. 56700/147) to HB. We would like to express our sincere gratitude to colleagues in Royan Institute, Regenerative Medicine Department.

Author information

Authors and Affiliations

Authors

Contributions

ZH performed the experiments, collected the data, conducted the data analysis and interpretation, and wrote the manuscript. IZ, MG, MA, SK, EZ, ZF, AP, IA, AS, RG, and PT helped in some experiments and analysis for the results, data management and statistical analysis and made final approval of the manuscript. HB and MV generated the concept, designed the study, supported the study and involved in writing and critical editing and proofreading the manuscript.

Corresponding author

Correspondence to Massoud Vosough.

Ethics declarations

Conflict of interest

The authors declare that there is no conflict of interest.

Ethical approval

This project was approved by Ethics Committee, Royan Institute. The approval code number is IR.ACECR.ROYAN.REC.1397.044. All institutional and national guidelines for the care and use of laboratory animals were followed.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 3137 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heydari, Z., Zarkesh, I., Ghanian, MH. et al. Biofabrication of size-controlled liver microtissues incorporated with ECM-derived microparticles to prolong hepatocyte function. Bio-des. Manuf. 4, 790–805 (2021). https://doi.org/10.1007/s42242-021-00137-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s42242-021-00137-4

Keywords

Navigation